AU2021236366A1 - Bcl-2 family proteins modulating compounds for cancer treatment - Google Patents

Bcl-2 family proteins modulating compounds for cancer treatment Download PDF

Info

Publication number
AU2021236366A1
AU2021236366A1 AU2021236366A AU2021236366A AU2021236366A1 AU 2021236366 A1 AU2021236366 A1 AU 2021236366A1 AU 2021236366 A AU2021236366 A AU 2021236366A AU 2021236366 A AU2021236366 A AU 2021236366A AU 2021236366 A1 AU2021236366 A1 AU 2021236366A1
Authority
AU
Australia
Prior art keywords
aryl
alkyl
cio
compound
groups
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
AU2021236366A
Inventor
Martina DE PASCALE
Camille DENIS
Nicolas GUEDENEY
Charline KIEFFER
Hippolyte PAYSANT
Laurent Poulain
Anne-Sophie Voisin-Chiret
Louis Bastien WEISWALD
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
CENTRE REGIONAL DE LUTTE CONTRE LE CANCER FRANCOIS BACLESSE
Universite de Caen Normandie
Institut National de la Sante et de la Recherche Medicale INSERM
Original Assignee
CENTRE REGIONAL DE LUTTE CONTRE LE CANCER FRANCOIS BACLESSE
Universite de Caen Normandie
Institut National de la Sante et de la Recherche Medicale INSERM
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by CENTRE REGIONAL DE LUTTE CONTRE LE CANCER FRANCOIS BACLESSE, Universite de Caen Normandie, Institut National de la Sante et de la Recherche Medicale INSERM filed Critical CENTRE REGIONAL DE LUTTE CONTRE LE CANCER FRANCOIS BACLESSE
Publication of AU2021236366A1 publication Critical patent/AU2021236366A1/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings

Landscapes

  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

The present invention relates to compounds, compositions and methods for treating cancers and disorders of cell proliferation and more particularly, methods of making and using compounds that modulate at least one of the proteins chosen from Bcl-2, Mcl-1, Bcl-X

Description

BCL-2 FAMILY PROTEINS MODULATING COMPOUNDS FOR CANCER TREATMENT
The present invention relates to compounds, compositions and methods for treating cancers and disorders of cell proliferation and more particularly, methods of making and using compounds that modulate at least one of the proteins chosen from Bcl-2, Mcl-1, Bcl- XL, Bcl-w, Bfl-1 and Bcl-B, in particular Bcl-2 protein, or BC1-XL and/or Mcl-1 proteins; said compounds may be contained in pharmaceutical compositions and used as therapeutic agents.
Cancer is a leading cause of death worldwide. Apoptosis, also known as programmed cell death, is a natural process used by multicellular organisms to eliminate aging or damaged cells also involved in various physiological processes such as morphogenesis and tissue homeostasis. Apoptosis is a complex, highly regulated process involving many proteins. Some of these proteins promote cell death ("pro-apoptotic" proteins) and some prevent it ("anti-apoptotic" proteins). Cancer cells tend to over-express anti-apoptotic genes. The over expression of anti-apoptotic genes is associated with tumor formation, metastatic growth and resistance to chemotherapy and there is a continuing need for therapeutic strategies that selectively kill cancer cells.
More specifically, apoptosis control defect is frequently involved in chemoresistance in cancer cells, in both hematological malignancies and solid tumors, and the deregulation of Bcl-2 family members expression constitutes one of the most frequent and important event. These proteins share Bcl-2 homology domains (named BH domains). Anti-apoptotic proteins (Bcl-2, BC1-XL...) contain the BH1 to BH4 domains, whereas pro-apoptotic proteins contain either the BH1 to BH3 domains (multidomain members such as Bax and Bak) or only the BH3 domain (BH3-only group such as Bim, Puma, Bid, Bad, Noxa and Hrk). Under cellular stress, BH3-only proteins initiate apoptosis by either blocking the activity of anti- apoptotic members or directly activating multidomain pro-apoptotic members, which is mediated via interaction of the BH3 domain of one protein with the hydrophobic pocket of another.
Constant efforts are made to impede the activity of anti-apoptotic members such as Bcl-2 or BC1-XL, among which the development of potent BH3-mimetic molecules represent a promising way. These molecules bind to the BH3-binding groove in anti-apoptotic proteins of the Bcl-2 family and promote cell death through the release of pro-apoptotic Bcl-2 family members. In ovarian carcinoma, inventors previously demonstrated that BC1-XL and Mcl-1 cooperate to protect tumor cells against apoptosis, and that their concomitant inhibition leads to massive apoptosis even in absence of chemotherapy, whereas the down-regulation of either BC1-XL or Mcl-1 may in some cases remain ineffective. In this context, they also showed that Mcl- 1 down regulation or inactivation was required to sensitize ovarian cancer cells to BCI-XL- targeting BH3-mimetic molecules such as ABT-737.
In hematological malignancies, including lymphomas, the interest of the targeting of Bcl-2 family anti-apoptotic proteins has been extensively documented. For instance, Bcl-2 has been successfully targeted in vitro as well as in vivo by the US FDA-approved BCL-2 inhibitor Venetoclax (or Venclaxta, developed by Abbvie) that has been found to be clinically active against various hematologic malignancies including multiple myeloma, acute myeloid leukemia, myelodysplastic syndrome, acute lymphoblastic leukemia, small lymphocytic lymphoma and others.
Other studies also showed that the inhibition of Mcl-1 improves the therapeutic efficiency of Bcl-2 or Bcl-xL-targeting BH3-mimetics in hematological malignancies. In other lymphoid malignancies, such as the aggressive mantle cell lymphoma (MCL), cell survival might rely instead or also on BCL-2 relative Mcl-1.
Mcl-1 contains three BH domains (BH1-BH3) but lacks a clearly defined BH4 domain at the NFb terminus. Mcl-1 localizes to various intracellular membranes, especially, the outer mitochondrial membrane through a transmembrane domain at its COOH terminus. Like Bcl-2 and BCI-XL, Mcl-1 can interact with Bak to inhibit mitochondria-mediated apoptosis. Unlike Bcl-2 and BCI-XL, Mcl-1 expression is quickly induced upon exposure to cytokines or growth factors. Increased Mcl-1 expression promotes cell viability in a wide range of tumor cell types, including leukemias, hepatocellular carcinomas, melanoma, prostate and breast cancer cells. Moreover, Mcl-1 plays a role in cell immortalization and tumorigenesis in many kinds of cancers through amplification of somatic copy number. Cancer cells harboring Mcl-1 amplification are frequently dependent upon Mcl-1 for survival.
Mcl-1 is overexpressed in various tumor cells, including ovarian carcinoma, hematological malignancies, breast cancers, colon cancers and lung cancers, and its expression has also been associated to chemoresistance. Importantly, Mcl-1 locus is one of the most frequently amplified in human cancers, further pointing to its centrality in carcinogenesis and increasing its importance as a high priority therapeutic target. However, the development of several molecules inhibiting the Mcl-1 pathway has been recently abandoned for reasons of cardiac toxicity.
Among the other promising therapeutic targets, the anti-apoptotic BC1-XL protein constitute a target of choice. This is particularly true in the management of certain solid tumors, which overexpress this protein (including ovarian tumors), and whose expression is strongly correlated with drug resistance. Although the pharmacological benefit of targeting this protein is not questioned at present, the development of molecules inhibiting this protein is currently hampered by the emergence of thrombocytopenia in patients.
Accordingly, it is an object of the present invention to provide alternative compounds useful for modulating Mcl-1 activity and/or BC1-XL, in particular degrading Mcl-1 and/or BC1-XL protein, while having no or low toxicity towards heart and platelets.
Another object of the present invention is to provide alternative compounds to treat cancer, notably hematologic malignancies, such as lymphomas, and/or solid tumors, such as ovarian cancers, while having no or low toxicity towards heart and platelets.
The present invention is thus directed, in one aspect, to a compound of following formula (I) :
Wherein:
Yi, Y2, Y3 and Y4 are each independently C or N, Yi and Y2 being such as one is N and the other is C, Y3 and Y4 being such as one is N and the other is C;
An, An are each independently selected from (C6-Cio)aryl, (5 to 7 membered) heteroaryl, -0-(C6-Cio)aryl, -0-(5 to 7 memberedjheteroaryl, -S-(C6-Cio)aryl, -S-(5 to 7 mem bcrcd) h ctcro ary l , -(Ci-C6)alkyl-(C6-Cio)aryl, -(Ci-C6)alkyl-(5 to 7 membered) heteroaryl and halogens, in particular Cl, said aryl and heteroaryl groups being optionally substituted by one to three R3 groups, i and j are independently 0 or 1, and
Ri, R2, are at each occurrence, independently selected from (Ci-C6)alkyl, (C6- Cio)aryl, -(Ci-C6)alkyl-(C6-Cio)aryl, -(C2-C6)alkenyl-(C6-Cio)aryl, -carbonyl-(C6-Cio)aryl-, -carbonyl-(Ci-C6)alkyl-(C6-Cio)aryl, C(=0)H, COOH, OH, said alkyl groups being optionally substituted by OH, said aryl groups being optionally substituted by one to three R4 groups; k and 1 are independently 0 or 1 ; at least one of (Ri)k and (R2)I being such as:
Ri is selected from (Ce-Cio)aryl, -(Ci-Ce)alkyl-(C6-Cio)aryl, -(C2-Ce)alkenyl- (C6-Cio)aryl, -carbonyl-(C6-Cio)aryl-, -carbonyl-(Ci-Ce)alkyl-(C6-Cio)aryl, andk is 1 ; or
R2 is selected from (C6-Cio)aryl, -(Ci-C6)alkyl-(C6-Cio)aryl, -(C2-C6)alkenyl- (C6-Cio)aryl, -carbonyl-(C6-Cio)aryl-, -carbonyl-(Ci-C6)alkyl-(C6-Cio)aryl, and 1 is 1;
R3 IS, at each occurrence, independently selected from (Ci-C6)alkyl, (Ci-C6)alkoxy, OH, C(=0)H, (CH2)mCONRaRb, C(=0)Rc, (CH2)„C02H, (CH2)„C02Rc, (CH2)PCN, (CH2)qC(=N(OH))NH2, -S02NRaRb, I, Cl, Br, F, (C6-Cio)aryl, a (5 to 7 membered)heteroaryl, -(Ci-C6)alkyl-(C6-Cio)aryl, -(C2-C6)alkenyl-(C6-Cio)aryl, said alkyl groups being optionally substituted by a OH, (Ci-C6)alkoxy and/or -0-(C6-Cio)aryl group;
R4 is, at each occurrence, independently selected from (Ci-C6)alkyl, (CH2)nC02H, (CH2)nC02Rc, I, Cl, Br, F, or there are two R4 groups that form with the aryl to which they are bound a naphthyl;
Ra and Rb are independently chosen from H and (C1-C6) alkyl;
Rc is chosen from H and (Ci-C6)alkyl; m is 0, 1,2,3 ; n is 0, 1,2,3 ; p is 0, 1,2,3 ; q is 0, 1,2,3 ; r is 0 or 1 ;
W and Y are each independently selected from a single bond, -CH=CH-, -CºC-, - NH-,-NRs-, -0-, -S-, -C(=0)NH-, -NHC(=0)-, -C(=0)0-, -OC(=0)-;
R5IS, at each occurrence, independently selected from (Ci-C6)alkyl,
Li and L2 are each independently selected from (Ci-Cis)alkyl, (C2-Cis)alkenyl, and (C2-Cis)alkynyl groups, said groups being optionally interrupted by one or more groups selected from -0-, -S-, -NR6-;
Reis, at each occurrence, independently selected from H and (Ci-Ce)alkyl, X is selected from (5 to 7 membered) heteroaryl, in particular a triazole, -NH- -NR7- , -0-, -S-, -C(=0)NH-, -NHC(=0)-, -C(=0)0-, -0C(=0)-;
R7IS selected from (Ci-C6)alkyl,
Z is a E3 ubiquitin ligase ligand; and the pharmaceutically acceptable salts thereof.
According to an embodiment, Yi and Y3 are N, and Y2 and Y4 are C.
According to an embodiment, Yi and Y3 are C, and Y2 and Y4 are N.
According to an embodiment, Yi and Y4 are N, and Y2 and Y34 are C.
According to an embodiment, when j is 0, then Y4 is N.
According to an embodiment, the compound of formula (I) as defined above is of following formula (II): (II).
According to an embodiment, the compound as defined above is of following formula (Ilia) or (Illb): said compound being in particular of following formula (Ilia 1 ) or (IIIa2), (Illbl) or
(IIIb2): (Ilia 1 )
In a particular aspect, there are included compounds as defined above wherein at least one of An, Ar2 is a (5 to 7 membered) heteroaryl, -0-(C6-Cio)aryl, -0-(5 to 7 membered)heteroaryl, -S-(C6-Cio)aryl, -S-(5 to 7 membered)heteroaryl, -(Ci-C6)alkyl-(C6- Cio)aryl, or -(Ci-C6)alkyl-(5 to 7 membered) heteroaryl, in particular a 5 to 7 membered heteroaryl containing a nitrogen atom, preferably pyridyl, notably 3-pyridyl.
In a particular aspect, there are included compounds as defined above wherein at least one of An, Ar2 is a (C6-Cio)aryl, in particular a phenyl.
According to an embodiment, An and/or Ar2 are selected from phenyl, pyridyl, pyrimidyl, imidazolyl, pyrazolyl, thiophenyl, triazolyl, in particular from phenyl, 3-pyridyl, 5-pyrimidyl, 2-imidazolyl, 3-pyrazolyl, 2-thiophenyl, 5-triazolyl.
According to an embodiment, An is selected from pyridyl, in particular from 3- pyridyl, j being notably 0.
According to an embodiment, An is 3-pyridyl or phenyl.
According to an embodiment, Ar2 is 3-pyridyl or phenyl.
According to an embodiment, W is selected from -CH=CH- and -CºC-, W being in particular -CºC-, j being notably O.According to an embodiment, Ri and R2 are independently selected from C1-C6 alkyl, in particular methyl, (C6-Cio)aryl, in particular phenyl or naphthyl, -(Ci-C6)alkyl-(C6-Cio)aryl, in particular -(Ci-C6)alkyl-phenyl, more particularly phenetyl, and -(Ci-C6)alkyl-naphthyl, more particularly ethyl-naphthyl , and - (C2-C6)alkenyl-(C6-Cio)aryl, in particular -(C2-C6)alkenyl -phenyl, more particularly styryl, and -(C2-C6)alkenyl-naphthyl, more particularly -vinyl-naphthyl.
According to a particular embodiment, Ri is selected from C1-C6 alkyl and R2 is selected from (C6-Cio)aryl, -(Ci-C6)alkyl-(C6-Cio)aryl, and -(C2-C6)alkenyl-(C6-Cio)aryl.
According to another particular embodiment, R2 IS selected from C1-C6 alkyl and Ri is selected from (C6-Cio)aryl, -(Ci-C6)alkyl-(C6-Cio)aryl, and -(C2-C6)alkenyl-(C6-Cio)aryl.
According to an embodiment, Ri and R2 are independently selected from C1-C6 alkyl and (C6-Cio)aryl(C2-C6)alkenyl, preferably from methyl, styryl and 2-(naphthalen-2- yl)vinyl.
According to a particular embodiment, Ri is selected from C1-C6 alkyl, in particular methyl and R2 is selected from -(Ci-C6)alkyl-(C6-Cio)aryl, in particular styryl or 2- (naphthalen-2-yl)vinyl.
According to another particular embodiment, R2 IS selected from C1-C6 alkyl and Ri is selected from (C6-Cio)aryl, -(Ci-C6)alkyl-(C6-Cio)aryl, and -(C2-C6)alkenyl-(C6-Cio)aryl.
According to an embodiment, Ri is 5-methyl.
According to an embodiment, R2 is 5-styryl or 2-(naphthalen-2-yl)vinyl.
According to another embodiment, R2 is 5-methyl.
According to another embodiment, Ri is 5-styryl or 2-(naphthalen-2-yl) vinyl.
In other aspects, there are included compounds of formula (I) wherein the pharmaceutically acceptable salts are hydrochloride, hydrobromide, hydroiodide, sulfate, nitrate, phosphate, acetate, citrate, propionate, fumarate, maleate, tartrate, glutamate, salicylate, oxalate, methanesulfonate, para-toluenesulfonate, succinate, or benzoate salts.
In other aspects, there are included compounds of formula (I) wherein the pharmaceutically acceptable salts are calcium, magnesium, lithium, potassium or sodium salts.
It is recognized that compounds of the present invention may exist in various stereoisomeric forms. As such, the compounds of the present invention include diastereomers, enantiomers and atropisomers. The compounds are normally prepared as racemates and can conveniently be used as such, but individual enantiomers can be isolated or synthesized by conventional techniques if so desired. Such racemates and individual enantiomers and mixtures thereof form part of the present invention.
It is well known in the art how to prepare and isolate such optically active forms. Specific stereoisomers can be prepared by stereo specific synthesis using enantiomerically pure or enantiomerically enriched starting materials. The specific stereoisomers of either starting materials or products can be resolved and recovered by techniques known in the art, such as resolution of racemic forms, normal, reverse-phase, and chiral chromatography, recrystallization, enzymatic resolution, or fractional recrystallization of addition salts formed by reagents used for that purpose. Useful methods of resolving and recovering specific stereoisomers described in Eliel, E. L.; Wilen, S.H. Stereochemistry of Organic Compounds ; Wiley: New York, 1994, and Jacques, J, et al. Enantiomers, Racemates, and Resolutions, Wiley: New York, 1981, each incorporated by reference herein in their entireties.
According to an embodiment, the compound as defined above is of following formula (IVa) or (IVb): in particular of following formula (IVal), (IVa2), (IVbl) or (IVb2): wherein :
Xi, X2, are at each occurrence, independently selected from C or N;
Ri, R2, R3 are independently, at each occurrence, as defined above.
According to an embodiment, the compound as defined above is of one of the following formulae:
or a stereoisomeric form, a mixture of stereoisomeric forms and a pharmaceutically acceptable salt form thereof, in particular a sodium or fumarate salt, wherein :
R3 and R4 are independently, at each occurrence, as defined above.
According to an embodiment, W is -NH-, Li is (Ci-Cis)alkyl, r is 1, X is a (5 to 7 membered) heteroaryl, in particular a triazole, L2 is a (Ci-Cis)alkyl interrupted by one or more -O- atoms, in particular of formula -CH2-(0-CH2-CH2)i-6- and Y is a single bond, - NH- or -C(=0)NH-.
According to another embodiment, W is -NH-, Li is a (Ci-Cis)alkyl interrupted by one or more -O- atoms, in particular of formula -CH2-CH2-(0-CH2-CH2)I-6-, r is 0 and Y is a single bond or -C(=0)NH-.
According to another embodiment, W is -CºC-, Li is a (Ci-Cis)alkyl interrupted by one or more -O- atoms, in particular of formula -CH2-(0-CH2-CH2)i-6-, r is 0 and Y is a single bond or -C(=0)NH-. According to an embodiment, Z is a VHL (von Hippel-Lindau), CRBN (cereblon), cIAP (Cellular Inhibitor of Apoptosis) or MDM2 (Mouse Double Minute 2) E3 ubiquitin ligase ligand, Z being in particular a VHL E3 ubiquitin ligase ligand.
According to a particular embodiment, Z is selected from:
In still other aspects, there are included compounds of formula (I) which are selected from:
Wherein :
R3 is H or as defined above, in particular Cl; Rs is (Ci-C6)alkyl, in particular propyl.
Method of preparation of compounds of formula (I)
The compounds of the present invention may be prepared in a number of methods well known to those skilled in the art, including, but not limited to those described below, or through modifications of these methods by applying standard techniques known to those skilled in the art of organic synthesis. The reagents and starting materials are commercially available, or readily synthesized by well-known techniques by one of ordinary skill in the arts. All substituents, unless otherwise indicated, are as previously defined. All processes disclosed in association with the present invention are contemplated to be practiced on any scale, including milligram, gram, multigram, or commercial industrial scale.
As will be readily understood, functional groups present on the compounds of Formula I may contain protecting groups. Protecting groups are known per se as chemical functional groups that can be selectively appended to and removed from functionalities, such as hydroxyl groups and carboxyl groups. These groups are present in a chemical compound to render such functionality inert to chemical reaction conditions to which the compound is exposed. Any of a variety of protecting groups may be employed with the present invention. Other preferred protecting groups according to the invention may be found in Greene, T.W. and Wuts, P.G.M., “Protective Groups in Organic Synthesis” 2d. Ed., Wiley & Sons, 1991, or in P.J. Kocienski, “Protecting Groups”, 3rd. Ed., Thieme, Stuttgart, NY, 2004.
The compound thus prepared may be recovered from the reaction mixture by conventional means. For example, the compounds may be distilled off from the solvent mixture after the extraction or, if necessary after distilling off from the solvent mixture, pouring the residue into water followed by extraction with a water-immiscible organic solvent and distilling off from the solvent mixture. Additionally, the product can, if desired, be further purified by various well known techniques, such as recrystallization, precipitation or the various chromatography techniques, notably column chromatography or preparative thin layer chromatography, in particular High Performance Liquid Chromatography (HPLC).
In another object, the present invention relates to a method for preparing a compound of formula (I) as defined herein, wherein r is 1, comprising the steps of: wherein Xa and Xb are chemical groups that form after chemical reaction the group X as defined above.
Xa and Xb are well known from the skilled person in the art and are for example -N3 and -= (alkynyl) respectively, forming a triazole by copper(I)-catalyzed azide-alkyne Huisgen cycloaddition.
Compound (B) is commercially available, as described below, or may be obtained as follows:
Xb-l-2— Ya + Yb-z - Xb-|_2-Y-z
(B1) (B2) (B) wherein Ya and Yb are chemical groups that form after chemical reaction the group Y as defined above.
Ya and Yb are well known from the skilled person in the art and are for example -COOH or an activated ester and -NH2 respectively, forming -CONH- by peptidic coupling, or -NH2 and -F (-F being in particular bound to an aromatic group), forming -NH- by nucleophilic aromatic substitution.
Compounds (Bl) and (B2) are commercially available, as described below or obtained by procedures well known from those skilled in the art.
In an embodiment, compound (A) is obtained as follows: wherein Wa and Wb are chemical groups that form after chemical reaction the group W as defined above.
Wa and Wb are well known from the skilled person in the art and are for example -F or -Cl and -NH2 respectively, forming -NH- by nucleophilic aromatic substitution, or - COOH or an activated ester and -NH2 respectively, forming -CONH- by peptidic coupling.
D is commercially available or obtained by procedures well known from those skilled in the art.
In an embodiment, compound (C) is obtained as follows, An being in particular selected from (C6-Cio)aryl and (5 to 7 membered) heteroaryl : when j is 1, and i is 0 or 1: wherein Hah is an halogen, in particular Br, or when j is 1, and i is 1: wherein Hali is an halogen, in particular Br, or when j=0, and i is 1: wherein Hali is an halogen, in particular Br.
When j=0 and i=0, compound (C) is then obtainable by procedures well known from those skilled in the art, in particular those described in WO2015132727.
When An is Hal, compound (C) may be obtained similarly to compound (G) as described below.
When An is selected from -0-(C6-Cio)aryl, -0-(5 to 7 membered)heteroaryl, -S-(C6- Cio)aryl, -S-(5 to 7 membered)heteroaryl, -(Ci-C6)alkyl-(C6-Cio)aryl, -(Ci-C6)alkyl-(5 to 7 membered) heteroaryl, compound (C) may be obtained as follows: for An = -0-(C6-Cio)aryl, -0-(5 to 7 membered)heteroaryl, -S-(C6-Cio)aryl, or - S-(5 to 7 membered)heteroaryl, from compound (G), for example with Hali=Br, in contact with Ar-SH or Ar-OH compound, by nuclear aromatic substitution; for An = -(Ci-C6)alkyl-(C6-Cio)aryl, -(Ci-C6)alkyl-(5 to 7 membered) heteroaryl, from compound (G), for example with Hali=Br, in particular by Negishi coupling, in presence of an Ar-alkyl-MgBr compound.
In an embodiment, compound (E) is obtained as follows, when i is 1:
Wherein Hali and Hah are halogens, for example I and Br, respectively.
J is obtainable by procedures well known from those skilled in the art, in particular those described in WO2015132727.
When i is 0, (E) is obtainable by procedures well known from those skilled in the art, in particular those described in WO2015132727.
Alternatively, compound (E) may be obtained as follows: wherein Hals is an halogen, in particular I, Hah being for example Br. In an embodiment, compound (G) is obtained as follows: wherein Hah is an halogen, in particular I, Hali being for example Br. Alternatively, compound (G) may be obtained as follows: wherein HaU is an halogen, in particular I, Hali being for example Br.
(L) is commercially available or obtained by procedures well known from those skilled in the art.
(M) is obtainable by procedures well known from those skilled in the art, in particular those described in WO2015132727.
(N) is obtainable by procedures well known from those skilled in the art, in particular those described in WO2015132727.
In another object, the present invention relates to a method for preparing a compound of formula (I) as defined herein, wherein r is 0, comprising the steps of:
Compound (B2) is as mentioned above.
Compound (A’) is as defined above as compound (A). Ya and Yb are well known from the skilled person in the art and are for example -COOH or an activated ester and -NH2 respectively, forming -CONH- by peptidic coupling, or -Nth and -F (-F being in particular bound to an aromatic group), forming -NH- by nucleophilic aromatic substitution.
All the substituents, for example the group R3, may be modified before, between, or after the steps described above. For example, the group R3=OH can be modified to R3=0- propyl by SN2 reaction.
When W is CºC, the W-containing fragment can for example be introduced to the aromatic scaffold by a Sonogashira coupling as well known from the skilled in the art.
In another object, the present invention relates to a pharmaceutical composition comprising a compound of formula (I) as defined above, in admixture with at least one pharmaceutically acceptable excipient or carrier.
All the embodiments mentioned above for the compounds of formula (I) apply here, alone or in combinations.
In an embodiment, the pharmaceutical composition defined above further comprises an additional anti-cancer agent, in particular chosen from anti-EGFR, anti-Map kinases and anti-Akt compounds. Other exemplary additional anticancer agents include carboplatin, paclitaxel, doxorubicin, topotecan, and functional derivatives of the foregoing.
As will be apparent to one of ordinary skill in the art, the specific formulations of said pharmaceutical composition can be selected based on the type of cancer being treated. The compositions of the invention can be formulated for administration to a patient with materials that improve their stability and/or provide for a controlled or sustained release in vivo. These pharmaceutical compositions can be prepared in a well known manner in the pharmaceutical art and, can be administered by a variety of routes, depending upon whether local or systemic treatment is desired and upon the area to be treated.
Administration may be topical (including skin, ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery), pulmonary (e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal, intranasal, epidermal and transdermal), ocular, oral or parenteral.
In particular, compounds and compositions of the invention are administered by oral administration or by intravenous administration.
The compounds and pharmaceutical compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral use, carriers which are commonly used include lactose and com starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried com starch. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
Methods for ocular delivery can include topical administration (eye drops), subconjunctival, periocular or intravitreal injection or introduction by balloon catheter or ophthalmic inserts surgically placed in the conjunctival sac. Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; or intracranial, e.g., intrathecal or intraventricular administration.
The pharmaceutical compositions can be formulated so as to provide controlled, quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.
The pharmaceutical compositions can also be formulated by techniques well known from those skilled in the art, for example by nanovectorization, or by using a micronized form.
Pharmaceutical compositions usually comprise pharmaceutically acceptable, inorganic or organic carriers, preservatives, solubilizers, stabilizers, wetting agents, emulsifiers, sweeteners, colorants, flavorings, salts for varying the osmotic pressure, buffers, masking agents or antioxidants. They can also contain still other therapeutically valuable substances. These ingredients are selected on the basis of the mode and route of administration. Suitable pharmaceutical ingredients, as well as pharmaceutical necessities for use in pharmaceutical formulations, are described in Remington's Pharmaceutical Sciences (E. W. Martin).
Pharmaceutically acceptable carriers that may be used in these pharmaceutical compositions include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as prolamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene- polyoxypropylene block polymers, polyethylene glycol and wool fat.The therapeutic dosage of the compounds of the present invention can vary according to, for example, the particular use for which the treatment is made, the manner of administration of the compound, the health and condition of the patient, and the judgment of the attending clinician. The proportion or concentration of a compound of the invention in a pharmaceutical composition can vary depending upon a number of factors including dosage, chemical characteristics (e.g., hydrophobicity), and the route of administration.
In a further object, the present invention thus relates to a compound of formula (I) as defined above for use in the treatment of cancer, notably of hematologic malignancies, such as lymphoma, leukemia, multiple myeloma, and/or of solid tumors such as ovarian cancers, mesothelioma, melanoma, pancrea, lung, breast, kidney and liver cancers.
All the embodiments mentioned above for the compounds of formula (I) apply here, alone or in combinations.
In a particular embodiment, the present invention thus relates to a compound of formula (I) as defined above for use in the treatment of cancer by inducing apoptosis mediated by at least one of the proteins chosen from Bcl-2, Mcl-1, BC1-XL, Bcl-w, Bfl-1 and Bcl-B, in particular of Bcl-2 protein, or BC1-XL and/or Mcl-1 proteins, more particularly of Bcl-2 protein, or BC1-XL and Mcl-1 proteins.
Cancers amenable to the therapeutic methods of the invention can be cancers that are responsive to the modulation of at least one of the proteins chosen from Bcl-2, Mcl-1, Bcl- XL, Bcl-w, Bfl-1 and Bcl-B, in particular of Bcl-2 protein, or BC1-XL and/or Mcl-1 proteins; any form of cancer which is associated with misregulation of at least one of the proteins chosen from Bcl-2, Mcl-1, BC1-XL, Bcl-w, Bfl-1 and Bcl-B, in particular of Bcl-2 protein, or BC1-XL and/or Mcl-1 proteins (e.g., overexpression or altered binding or activity) is thus within the scope of the invention.
Cancers or neoplastic disorders include, for example, without limitation, breast cancer, hematological cancers such as myeloma, leukemia and lymphoma (e.g., Burkitt lymphoma, non-Hodgkin lymphoma, Hodgkin lymphoma, and acute T cell leukemia), neurological tumors such as brain tumors, e.g., gliomas, including astrocytomas or glioblastomas, melanomas, lung cancer, head and neck cancer, thyroid cancer, gastrointestinal tumors such as stomach, colon or rectal cancer, liver cancer, pancreatic cancer, genitourinary tumors such ovarian cancer, vaginal cancer, vulval cancer, endometrial cancer, bladder cancer, kidney cancer, testicular cancer, prostate cancer, or penile cancer, bone tumors, vascular tumors, and skin cancers such as basal cell carcinoma, squamous cell carcinoma and melanoma.
In another aspect, there are included compounds of formula (I), for use as a ligand of at least one of the proteins chosen from Bcl-2, Mcl-1, BC1-XL, Bcl-w, Bfl-1 and Bcl-B, in particular of Bcl-2 protein, or BC1-XL and/or Mcl-1 proteins, more particularly of Bcl-2 protein, or BC1-XL and Mcl-1 proteins.
In another aspect, there are included compounds of formula (I), for use for inducing apoptosis mediated by at least one of the proteins chosen from Bcl-2, Mcl-1, BC1-XL, Bcl-w, Bfl-1 and Bcl-B, in particular of Bcl-2 protein, or BC1-XL and/or Mcl-1 proteins, more particularly of Bcl-2 protein, or BC1-XL and Mcl-1 proteins.
A patient is effectively treated whenever a clinically beneficial result ensues. This may mean, for example, a complete resolution of the symptoms of a disease, a decrease in the severity of the symptoms of the disease, or a slowing of the disease's progression.
An effective amount of any composition provided herein can be administered to an individual in need of treatment. The term "effective" as used herein refers to any amount that induces a desired response while not inducing significant toxicity in the patient. Such an amount can be determined by assessing a patient's response after administration of a known amount of a particular composition.
In another aspect, the inventions relate to a method of treating cancer comprising administering to a patient in need thereof an effective amount of pharmaceutical composition comprising a compound of formula (I) as defined above. In another aspect, the inventions relate to a method of degrading at least one of the proteins chosen from Bcl-2, Mcl-1, BC1-XL, Bcl-w, Bfl-1 and Bcl-B, in particular of Bcl-2 protein, or BCI-XL and/or Mcl-1 proteins, more particularly of Bcl-2 protein, or BCI-XL and Mcl-1 proteins, comprising contacting said protein with a compound of formula (I) as defined above.
In another aspect, the inventions relate to a method of degrading at least one of the proteins chosen from Bcl-2, Mcl-1, BCI-XL, Bcl-w, Bfl-1 and Bcl-B, in particular Bcl-2 protein, or BCI-XL and/or Mcl-1 proteins, more particularly Bcl-2 protein, or BCI-XL and Mcl- 1 proteins, in vivo comprising contacting the protein with a compound of formula (I) as defined above to degrade said protein by ubiquitination targeted by said compound of formula (I).
The present invention also relates to methods of killing cancer cells and methods of modulating levels of at least one of the proteins chosen from Bcl-2, Mcl-1, BCI-XL, Bcl-w, Bfl-1 and Bcl-B, in particular of Bcl-2 protein, or BCI-XL and/or Mcl-1 proteins in a cell. The therapeutic methods described herein can be carried out in connection with other cytotoxic therapies (e.g., chemotherapy, hormone therapy, radiotherapy, antibody-based therapies or targeted therapies directed against cell signaling pathways).
In a preferred embodiment, the compound and the composition of the invention are useful for preventing or reducing metastasis or further dissemination in patient suffering from Bcl-2, Mcl-1, BCI-XL, Bcl-w, Bfl-1 and Bcl-B, in particular Bcl-2, or BCI-XL and/or Mcl-1 expressing cancer; more specifically, they are useful for increasing the duration of survival of such a patient, increasing the progression free survival of such a patient, increasing the duration of response, resulting in a statistically significant and clinically meaningful improvement of the treated patient as measured by the duration of survival, progression free survival, response rate or duration of response. In a preferred embodiment, the medicament is useful for increasing the response rate in a group of patients.
Definitions
The following terms and expressions contained herein are defined as follows:
As used herein, the term “alkyl” refers to a straight-chain, or branched alkyl group having 1 to 8 carbon atoms, such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec- butyl, tert-butyl, pentyl, isoamyl, neopentyl, 1-ethylpropyl, 3-methylpentyl, 2,2- dimethylbutyl, 2,3-dimethylbutyl, hexyl, octyl, etc. The alkyl moiety of alkyl-containing groups, such as alkoxy, and alkoxycarbonyl, has the same meaning as alkyl defined above. Lower alkyl groups, which are preferred, are alkyl groups as defined above which contain 1 to 4 carbons. A designation such as “C1-C6 alkyl” refers to an alkyl radical containing from 1 to 6 carbon atoms.
As used herein, the term “alkenyl” refers to a straight chain, or branched hydrocarbon chains of 2 to 6 carbon atoms having at least one carbon-carbon double bond. A designation “C2-C6 alkenyl” refers to an alkenyl radical containing from 2 to 6 carbon atoms. Examples of alkenyl groups include ethenyl, propenyl, isopropenyl, 2,4-pentadienyl. “C2-C4 alkenyl” are particularly preferred.
As used herein, the term “alkoxy” means an alkyl-O- group wherein the alkyl group is as herein described. Exemplary alkoxy groups include methoxy, ethoxy, n-propoxy, i- propoxy, and n-butoxy.
As used herein, the term “aryl” refers to a substituted or unsubstituted, mono- or bicyclic hydrocarbon aromatic ring system having 6 to 10 ring carbon atoms. Examples include phenyl and naphthyl.
As used herein, the term “arylalkyl” refers to an alkyl group that is substituted with an aryl group. Examples of arylalkyl groups include, but are not limited to, benzyl, bromobenzyl, phenethyl, benzhydryl, diphenylmethyl, triphenylmethyl, diphenylethyl, naphthylmethyl.
As used herein, the term “arylalkenyl” refers to an alkenyl group that is substituted with an aryl group. Examples of arylalkenyl include, but are not limited to, styryl.
As used herein, the term “arylcarbonyl” refers to an aryl-C(=0)- group wherein the aryl group is as herein described.
As used herein, the term “arylalkylcarbonyl” refers to an arylalkyl-C(=0)- group wherein the arylalkyl group is as herein described.
As used herein, the term “heteroaryl” refers to an aromatic group containing 5 to 10 ring carbon atoms, preferably 5 to 7, in which one or more ring carbon atoms are replaced by at least one hetero atom such as -0-, -N-, or -S-. Examples of heteroaryl groups include pyrrolyl, furanyl, thienyl, pyrazolyl, imidazolyl, thiazolyl, isothiazolyl, isoxazolyl, oxazolyl, oxathiolyl, oxadiazolyl, triazolyl, oxatriazolyl, furazanyl, tetrazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, indolyl, isoindolyl, indazolyl, benzofuranyl, isobenzofuranyl, purinyl, quinazolinyl, quinolyl, isoquinolyl, benzoimidazolyl, benzothiazolyl, benzothiophenyl, thianaphthenyl, benzoxazolyl, benzisoxazolyl, cinnolinyl, phthalazinyl, naphthyridinyl, and quinoxalinyl. Included within the definition of “heteroaryl” are fused ring systems, including, for example, ring systems in which an aromatic ring is fused to a heterocycloalkyl ring. Examples of such fused ring systems include, for example, phthalamide, phthalic anhydride, indoline, isoindoline, tetrahydroisoquinoline, chroman, isochroman, chromene, and isochromene.
As used herein, the term “subject” refers to a warm blooded animal such as a mammal, preferably a human, or a human child, which is afflicted with, or has the potential to be afflicted with one or more diseases and conditions described herein.
As used herein, a “therapeutically effective amount” refers to an amount of a compound of the present invention effective to prevent or treat the symptoms of particular disorders. Such disorders include, but are not limited to, those pathological and neurological disorders associated with the aberrant activity of the receptors described herein, wherein the treatment or prevention comprises inhibiting, inducing, or enhancing the activity thereof by contacting the receptor with a compound of the present invention.
As used herein, the term “pharmaceutically acceptable” refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem complications commensurate with a reasonable benefit/risk ratio.
The meanings of all the other terms used in the description of the present invention are well known in the art.
In another aspect, the present invention is directed to pharmaceutically acceptable salts of the compounds described above. As used herein, “pharmaceutically acceptable salts” includes salts of compounds of the present invention derived from the combination of such compounds with non-toxic acid or base addition salts.
Acid addition salts include inorganic acids such as hydrochloric, hydrobromic, hydroiodic, sulfuric, nitric and phosphoric acid, as well as organic acids such as acetic, citric, propionic, fumaric, maleic, tartaric, glutamic, salicylic, oxalic, methanesulfonic, para- toluenesulfonic, succinic, and benzoic acid, and related inorganic and organic acids.
Base addition salts include those derived from inorganic bases such as ammonium and alkali and alkaline earth metal hydroxides, carbonates, bicarbonates, and the like, as well as salts derived from basic organic amines such as aliphatic and aromatic amines, aliphatic diamines, hydroxy alkamines, and the like. Such bases useful in preparing the salts of this invention thus include ammonium hydroxide, potassium carbonate, sodium bicarbonate, calcium hydroxide, methylamine, diethylamine, ethylenediamine, cyclohexylamine, ethanolamine and the like.
In addition to pharmaceutically acceptable salts, other salts are included in the invention. They may serve as intermediates in the purification of the compounds, in the preparation of other salts, or in the identification and characterization of the compounds or intermediates.
FIGURES
Figure 1 presents the CRBN expression on IGROV1-R10 ovarian cancer cell line, according to example 4.
Figure 2 illustrates the expression of Mcl-1 and BCI-XL after a 24h exposure of IGROV1- R10 cells to 1 mM Pyridoclax or to 1 mM CRBN-PBMl-001 quantified by densitometry analysis of the western blot signals, according to example 4.
EXAMPLES
Example 1: Synthesis of compounds of formula (I)
Material and method are described below.
Material
Commercial reagents were used as received without additional purification. Melting points were determined on a Kofler heating bench. IR spectra were recorded on a Perkin Elmer BX FT-IR spectrophotometer. The band positions are given in reciprocal centimeters (cm 1). 1 H NMR (400 MHz) and 13C NMR (100 MHz) spectra were recorded on a JEOL Lambda 400 spectrometer. Chemical shifts are expressed in parts per million downfield from tetramethylsilane as an internal standard and coupling constants in Hertz. Chemical shift are reported in part per million (ppm) relative to the solvent resonance. Chromatography was carried out on a column using flash silica gel 60 Merck (0.063-0.200 mm) as the stationary phase. The eluting solvent indicated for each purification was determined by thin layer chromatography (TLC) performed on 0.2 mm precoated plates of silica gel 60F-264 (Merck) and spots were visualized using an ultraviolet-light lamp. Elemental analyses for new compounds were performed and the data for C, H, and N were within ± 0.4 of the theoretical values for all final compounds.
Methods Compounds of the invention were synthesized as follows. Reactions were performed in the order indicated in brackets, according to the general procedures mentioned below:
2-[2-(2-propynyloxy)ethoxy]ethylamine Fluorochem 519527
2-(2,6-Dioxo-piperidin-3-yl)-4-fluoroisoindoline-1,3- dione
Fluorochem 544654
2-[2-(2-propynyloxy)ethoxy]ethylamine Fluorochem 519527
- General procedure for Suzuki-Miyaura cross-coupling reaction: To a reaction vessel containing a solution of the appropriate bromo- or iodopyridine derivative in 1,4-dioxane (0.2 M) under inert atmosphere, the boronic acid reactant (1.1 equiv), tetrakis(triphenylphosphine)palladium(0) (5 mol%) and saturated K3PO4 aqueous solution (2.5 equiv) were added. The mixture was refluxed until completion of the reaction (monitored by TLC). The mixture was cooled to room temperature, quenched with water and extracted in ethyl acetate. The organic layers were collected, washed with brine, dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure to give the crude residue which was purified by column chromatography.
- General procedure for Nucleophilic Aromatic Substitution SN,\·:
The amine was dissolved in dry DMSO and DIPEA (N,N-diisopropylethylamine) (2 to 4 equiv). Fluoro- or chloro(pyridyl) or (benzyl) compound (1.0 equiv) was added, and the mixture was stirred at 90 °C until completion of the reaction (monitored by TLC). After cooling to rt, the mixture was poured onto half- saturated brine and it was extracted with EtOAc. The combined organic layers were further washed with saturated NH4CI solution, 5% LiCl solution, and brine. The organic layers were dried over NaiSCE, filtered and concentrated in vacuo.
- General procedure for Nucleophilic Substitution SN2: To a reaction vessel containing phenol compound (1.0 equiv) in DMF, iodoalkane (2.0 equiv) and K2CO3 (4.0 equiv) were added at 0 °C. The reaction mixture was stirred at rt until completion of the reaction (monitored by TLC). The mixture was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure to give the crude residue which was purified by column chromatography . - General procedure for copper(I)-catalyzed azide-alkyne cycloaddition CuACC:
To a mixture of azide compound (1.0 equiv) and alkyne compound (1.33 equiv) in 'BuOH- THF (1:1, v/v) under inert atmosphere was added CuS04-5H20 (2.2 equiv) and sodium L- ascorbate (2.2 equiv) in water. The mixture was stirred at 55 °C until completion of the reaction (monitored by TLC) and cooled to room temperature. Then it was poured into water and extracted with DCM. The organic phase was washed with brine, dried over NaiSC , filtered and evaporated to dryness. The crude product was purified by silica gel flash column chromatography using DCM and MeOH as eluents.
- General procedure for nitrile hydrolysis:
Benzonitrile compound (1.0 equiv) was stirred in a mixture of NaOH (2N) and MeOH at 100 °C until completion of the reaction (monitored by TLC). The reaction mixture was cooled at rt. The reaction mixture was acidified by AcOH until the apparition of a precipitate. The precipitate was filtered.
- General procedure for peptidic coupling reaction:
The carboxylic acid compound (1.0 equiv.) was dissolved in dry DMF and DIPEA (3.0 equiv) and HATU (1.1 equiv) were added under inert atmosphere. After stirring for 5 min, the amine derivative (1.1 equiv.), dissolved in dry DMF was added. The combined mixture was stirred at rt until completion of the reaction (monitored by TLC). Half-saturated brine was added and it was extracted with EtOAc. The combined organic layers were further washed with saturated NH4CI solution, 5% LiCl solution, and brine. The organic layers were dried over NaiSCC, filtered and concentrated in vacuo.
- General procedure for Boc-deprotection and HATU coupling:
The Boc-protected linker conjugate (1.0 equiv) was dissolved in dry CH2CI2 and it was treated with trifluoroacetic acid. The mixture was stirred until completion of the reaction at rt. The solvent was removed and it was co-evaporated with dry CH2CI2. The oily residue was further dried in high vacuum. The deprotected acid derivative (1.0 equiv) was dissolved in dry DMF and DIPEA (3.0 equiv) and HATU (1.1 equiv) were added. After stirring for 5 min, the amine derivative (1.1 equiv) dissolved in dry DMF, was added. The combined mixture was stirred at rt until completion of the reaction (monitored by TLC). Half- saturated brine was added and it was extracted with EtOAc. The combined organic layers were further washed with saturated NH4CI solution, 5% LiCl solution, and brine. The organic layers were dried over NaiSCE, filtered and concentrated in vacuo.
Starting compounds mentioned above have been obtained as follows: analogously to what has been described in WO2015132727.
When An is selected from -0-(C6-Cio)aryl, -0-(5 to 7 membered)heteroaryl, -S-(C6- Cio)aryl, -S-(5 to 7 membered)heteroaryl, -(Ci-C6)alkyl-(C6-Cio)aryl, -(Ci-C6)alkyl-(5 to 7 membered) heteroaryl, the An group may be introduced analogously as indicated below:
Kev: Nucleophilic Aromatic Substitution (SNAr)
Ullman-type coupling reaction (Ullman)
Palladocatalyzed Negishi cross coupling reaction (Negishi)
General procedure for Nucleophilic Aromatic Substitution SNAr: To a reaction vessel containing (E)-6-bromo-5-methyl-3'-styryl-3,2':5',3"-terpyridine (1.0 equiv) in DMSO, benzenethiol (1.0 equiv) and K2CO3 (2.0 equiv) were added. The reaction mixture was stirred at 110 °C for 48 h, obtaining only 50% conversion of starting material. Benzenethiol (1.0 equiv) and K2CO3 (2.0 equiv) were recharged in the reaction mixture and reaction was completed after 24h. The mixture was cooled to room temperature, quenched with water and extracted with DCM. The combined organic layers were washed with brine, dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure to give the crude residue which was purified by column chromatography (DCM/MeOH 95/5 to 9/1). General procedure for Ullman-type coupling reaction: To a reaction vessel containing (£')-6-bromo-5-methyl-3'-styryl-3,2':5',3"-terpyridine (1.0 equiv) in DMF, phenol (1.0 equiv), copper(0) powder (10%) and CS2CO3 (3.0 equiv) were added. The reaction mixture was stirred at 100 °C for 24 h. The mixture was cooled to room temperature, quenched with NaOH 1 M solution and extracted with DCM. The combined organic layers were washed with brine, dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure to give the crude residue which was purified by column chromatography (DCM/MeOH 98/2).
General procedure for palladocatalyzed Negishi cross coupling reaction: To a reaction vessel containing (£')-6-bromo-5-methyl-3'-styryl-3,2':5',3"-terpyridine (1.0 equiv) in dry THF under nitrogen atmosphere, benzylzinc(II) bromide 0.5 M (1.0 equiv) and tetrakis(triphenylphosphine)palladium(0) (10%) were added. The reaction mixture was stirred at rt for 48 h. The mixture was concentrated under reduced pressure to give the crude residue which was purified by column chromatography (DCM/MeOH 99/1 to 9/1).
Example 2 : Biological activity of compounds of formula (I)
Morphological observation of the cell nuclei
After treatment, both detached and adherent cells are pooled after trypsinization, applied to a polylysine-coated glass slide by cytocentrifugation and fixed with a solution of ethanol/chloroform/acetic acid (6:3:1). The preparations are then incubated for 15min at room temperature with lpg/ml DAPI solution (Boehringer Mannheim-Roche, Mannheim, Germany), washed in distillated water, mounted under a covers lip in Mowiol (Calbiochem) and analyzed under a fluorescence microscope (BX51, Olympus, Rungis, France). Condensed and fragmented nuclei indicate the induction of apoptosis.
Target expression and apoptosis induction analysis by western blot
Cells are rinsed with ice-cold PBS, suspended in a lysis buffer [RIPA : NaCl 150mM, Tris (pH 8) 50mM, Triton X100 1%, PMSF 4mM, EDTA 5mM, NaF lOmM, NaPPi lOmM, Na30V4 ImM, aprotinin 0.5pl/ml and 4.6 ml ultra-pure water] and incubated on ice for 30min. Lysates are collected after centrifugation (13200g, 10 min, 4°C) and protein concentrations are determined using the Bradford assay (Bio-Rad, Hercules, USA). Equal amounts of protein (20pg) are separated by SDS-PAGE on a 4- 15% gradient polyacrylamide Mini-PROTEAN® TGX™ precast gel (Bio-Rad) and transferred to PDVF membranes (Bio- Rad). Membranes are blocked lh at room temperature with 5% (v/v) non-fat dry milk in TBS with 0.05% (v/v) Tween20 (T-TBS). Membranes are then incubated overnight at 4°C with the appropriate antibodies, listed above. Membranes are then washed with T-TBS and incubated for 1 hour with the appropriate secondary antibody. Signals are revealed using Enhance ChemiLuminescence substrate (ECL) Prime Western Blot detection reagent (GE Healthcare Life Sciences) and the ImageQuant® Las4000Series (GE Healthcare Life Sciences), and then quantified by pixel densitometry using the ImageJ® software. Antibodies used: anti-Mcl-1 (#5453), anti-Bcl-XL (#2764), anti-Caspase 3 (#9662), anti- PARP (#9542), (Cell Signaling Technology), anti-Actin (#MAB1501) (Merck Millipore) and anti-VHL (#GTX101087) (Genetex)
Apoptotic events quantification using flow cytometry Adherent and floating cells are pooled, washed with phosphated-buffered saline (PBS) and fixed with ethanol 70%. Cells are then centrifuged at 2000r.p.m for 5min and incubated for 30min at 37°C in PBS, to allow the release of low molecular weight DNA. Cell pellets are stained with propidium iodide using the DNA Prep Coulter Reagent Kit (Beckman Coulter). Samples are analyzed using Gallios flow cytometer (Beckman Coulter) and cell cycle distribution is determined using Gallios software (Beckman-Coulter).
Real time cell activity analysis (impedancemetry, xCELLigence technology)
Real-time growth curves monitoring is performed with the Real-Time Cell Analyzer Multi- Plate instrument, using the xCELLigence System (ACEA, Ozyme, Saint Quentin en Yvelines, Lrance). This system monitors cellular events in real-time by measuring electrical impedance across interdigitated micro-electrodes integrated into the bottom surfaces of 96- well E-plates VIEW. These electrodes measure Cl (Cell Index) based on impedance. Cl correlates with the area of cells attached to the bottom of the plate. The Cl values are displayed in the plot. Briefly, cells are plated in 96-well E-Plate View and placed onto the RTCA MP located inside a tissue culture incubator. The cells are left to grow for 24 h before treatment and impedance is continuously measured until the end of the treatment. Standard deviations of wells triplicates are analyzed with the RTCA 2.1.0 Software
Real time cell imaging
Cells are seeded in 96-well plates in media. Cells are maintained at 37°C and 5% C02 and monitored using an IncuCyte® S3 (Essen BioScience). IncuCyte® Caspase-3/7 Green Apoptosis Assay Reagent (Essen Bioscience) is added the next day following treatment and baseline images are taken using lOx objective. Fluorescent and phase-contrast images are acquired in real time every 1 hour from 2 separate regions per well. The Caspase 3/7 reagent labels dead cells yielding green fluorescence. The live-cell phase contrast images are used to calculate confluence using the IncuCyte® software, and to provide morphology information. Each experiment is done in triplicate and accumulation of caspase 3/7 over time is normalized to confluence of cells.
E3 ligases expression characterization by immunohistochemistry
Immunohistochemistry using a Ventana Discovery XT autostainer is performed on 4 pm- thick paraffin sections. Slides are deparaffinized with EZPrep buffer and epitopes are unmasked by 15 min of high-temperature treatment in CC1 EDTA buffer. Sections are incubated for 40 min at 37°C with an antibody directed against various E3 ligases, among which anti VHL (GTX101087) (Genetex). Secondary antibody (Omnimap Rabbit; Ventana Medical System Inc., Tucson, AZ, USA) is incubated for 16 min at room temperature. Immunodetection performed without the primary antibody is used as the control. After washes, the staining is performed with DAB and sections are counterstained with hematoxylin using Ventana reagents according to the manufacturer's protocol. Stained slides are then digitized using an Aperio ScanScope slide scanner (Aperio Technologies, Vista, CA, USA).
Protein quantitation by ELISA
The colorimetric ELISA assays allows quantitative analysis on target protein expression. A primary antibody targets the protein of interest. A second antibody recognizes the primary and conjugated to HRP allows a colorimetric detection. The kits used for ELISA assay can be OKAG00550 for BC1-XL and OKAG00858 for Mcl-1, and procedures are applied according to the manufacturer's recommendations.
Results
Compounds of the invention induce proteolysis of Bcl-2, Mcl-1, BC1-XL, Bcl-w, Bfl-1 or Bcl- B protein, in particular of Bcl-2 protein, or Bcl-xL and/or Mcl-1 proteins.
Without being bound to any particular theory, it is believed that they recruit an E3 ligase to said target protein, which results in ubiquitination and subsequent degradation of the target protein by the proteasome.
Example 3: Synthesis of other compounds of the invention
As depicted below, in certain exemplary embodiments, compounds are prepared according to the following general procedures. It will be appreciated that, although the general methods depict the synthesis of certain compounds of the present invention, the following general methods, and other methods known to one of ordinary skill in the art, can be applied to all compounds and subclasses and species of each of these compounds, as described herein.
General Synthetic Methods The following examples are intended to illustrate the invention and are not to be construed as being limitations thereon. Temperatures are given in degrees centigrade. If not mentioned otherwise, all evaporations are performed under reduced pressure, preferably between about 15 mm Hg and 100 mm Hg (=20-133 mbar). The structure of final products, intermediates and starting materials is confirmed by standard analytical methods, e.g., microanalysis and spectroscopic characteristics, e.g., MS, IR, NMR. Abbreviations used are those conventional in the art.
All starting materials, building blocks, reagents, acids, bases, dehydrating agents, solvents, and catalysts utilized to synthesis the compounds of the present invention are either commercially available or can be produced by organic synthesis methods known to one of ordinary skill in the art. Further, the compounds of the present invention can be produced by organic synthesis methods known to one of ordinary skill in the art as shown in the following examples.
All reactions are carried out under nitrogen or argon unless otherwise stated.
Analytical instruments
All solvents and chemicals were used as purchased unless stated otherwise. All NMR spectra were recorded on Bruker Avance III 400 MHz or Avance III HD 500 MHz spectrometers. 1 H- and 13C NMR spectra are reported as chemical shifts (d) in parts per million (ppm). Coupling constants (J) are reported in hertz (Hz) to the nearest one decimal point. The following abbreviations are used to describe multiplets: s (singlet), d (doublet), t (triplet), q (quartet), m (multiplet), br (broad). Infrared spectra were recorded as neat compound using a Perkin Elmer FT-IR spectrometer. Absorptions are reported in wavenumbers (cm 1) and only peaks of interest are reported. Melting points of solids were measured with a Griffin apparatus. LC-MS was performed using a Waters SQ Detector as Mass Spectrometer and Waters Alliance 2695 as separating module (column XBridge C18 2.5 mM/4.6 50 mM) using the following gradients: A (H2O with 0.1% of HCChHyB (CH3CN with 0.1% of HCO2H) 95/5 to A/B 5/95 in 4.00 min. This ratio was held for 1.50 min before returning to the initial conditions for 0.50 min. Initial conditions were then maintained for 2.00 min. The retention times (tR) of the molecules analysed are indicated in min. Unless otherwise specified, all reagents were obtained from commercial suppliers. Chromatography was carried out using flash silica gel 60 Merck (0.063-0.200 mm) as the stationary phase. Flash chromatography were performed using a Biotage Isolera One flash chromatography. The eluting solvent, indicated for each purifications, was determined by thin layer chromatography (TLC) performed using Merck 0.2 mm precoated plates of silica gel with Kieselgel 60 F264. The products were visualized using UV fluorescence (254 nm).
Building block Synthetic Methods
5-Bromo-3-iodopyridin-2-ol 1. To a round bottom flask containing 5-bromopyridin-2-ol (5.0 g, 28.7 mmol) in acetonitrile (120 mL),/V-iodosuccinimide (7.1 g, 31.6 mmol, 1.1 equiv) was added. The solution was refluxed for 4 h, following the reaction by TLC. The suspension was cooled to room temperature and the solid was filtered and washed with MeOH to afford 7.1 g of 1 as a pink solid (83% yield).
Chemical Formula: CsHiBrINO ; Molecular Weight: 299.89 g/mol ; Melting Point: 63
°C
IR (KBr disc): 3490, 1630, 1607, 1522, 1457, 1309, 863, 683, 628 cm 1.
¾ NMR (CDCb) d 8.14 (d, /= 2.5 Hz, 1H), 7.59 (d, /= 2.5 Hz, 1H), 4.20 (br s, 1H, OH). 13C NMR (CDCb) d 171.5, 146.8, 145.9, 100.7, 92.9.
MS (ESI+): m/z 300.25 [M + H+], 302.26 [M+ + 2]
5-Bromo-2-chloro-3-iodopyridine 2. 5-Bromo-3-iodopyridin-2-ol 1 (9.0 g, 30.0 mmol) was dissolved in phenylphosphonic dichloride 90% (100 mL). The mixture was stirred and heated at 160 °C for 4 h, followed by TLC. The mixture was cooled down to room temperature and quenched by slow addition into a 1 L flask filled with ice and ammonia 28% (200 mL). The mixture was extracted with ethyl acetate, the organic layers were collected, washed with brine, dried over anhydrous magnesium sulfate, filtered, and concentrated under reduced pressure to give the crude residue which was purified by column chromatography (c-hexane/ethyl acetate 9/1) to afford 7.8 g of 2 as a white solid (82% yield). Chemical Formula: CsthBrClIN ; Molecular Weight: 318.34 g/mol ; Melting Point: 67 °C
IR (KBr disc): 3081, 3038, 1525, 1391, 1352, 1136, 1012, 890, 711, 515 cm-1.
!H-NMR (CDCb) d 8.40 (d, /= 2.3 Hz, 1H), 8.29 (d, /= 2.3 Hz, 1H).
13C-NMR (CDCb) d 153.2, 150.2, 149.6, 118.4, 95.3.
MS (ESI+): m/z 318.17 [M + H+], 320.14 [M+ + 2], 322.15 [M+ + 4]
(/?)-5-Bromo-2-chloro-3-styrylpyridine 3. To a reaction vessel containing 5-bromo-2- chloro-3-iodopyridine 2 (13.0 g, 40.8 mmol) in n-butanol (300 mL) under nitrogen atmosphere, (L)-styrylboronic acid (9.0 g, 61.2 mmol, 1.5 equiv), Pd(dppf)Cl2-DCM (166 mg, 0.21 mmol, 0.5 mol%) and K2CO3 (11.3 g, 81.6 mmol, 2.0 equiv) were added. The mixture was stirred at reflux overnight. The mixture was cooled to room temperature, quenched with water and extracted with ethyl acetate. The organic layers were collected, washed with brine, dried over anhydrous magnesium sulfate, filtered and then, concentrated under reduced pressure to give the crude residue which was purified by column chromatography (c-hexane/ethyl acetate 99/1, then 98/2) to afford 9.6 g of 3 as a white solid (80% yield).
Chemical Formula: Ci3H9BrClN ; Molecular Weight: 294.58 g/mol ; Melting Point: 104 °C
IR (KBr disc): 3036, 1627, 1530, 1494, 1447, 1389, 1152, 1116, 1063, 956, 897, 755, 719, 687, 491 cm-1.
3H-NMR (CDCb) d 8.35 (d, / = 2.5 Hz, 1H, HPyr), 8.11 (d, / = 2.5 Hz, 1H, HPyr), 7.57 (d, / = 7.4 Hz, 2H, HAT), 7.42 (t, / = 7.4 Hz, 2H, HAT), 7.38-7.34 (m, 1H, HAT), 7.27 (d, / = 16.3 Hz, 1H, Ar-CH=C7/-Pyr), 7.12 (d, / = 16.3 Hz, 1H, Ar-C//=CH-Pyr).
13C-NMR (CDCb) d 148.6, 148.5, 136.7, 135.9, 134.6 (Ar-CH=CH-Pyr), 133.7, 129.1, 128.9 (2 CATH), 127.1 (2 CATH), 121.9 (Ar-CH=CH-Pyr), 119.3.
MS (ESI+): m/z 294.33 [M + H+], 296.30 [M+ + 2], 298.30 [M+ + 4]
(£')-6-Cloro-5-styryl-3,3'-bipyridine 4. To a reaction vessel containing (£)-5-bromo-2- chloro-3-styrylpyridine 3 (3.8 g, 12.7 mmol) in 1,4-dioxane under nitrogen atmosphere, pyridin-3-ylboronic acid (1.7 g 14.0 mmol, 1.1 equiv), tetrakis(triphenylphosphine)palladium(0) (734 mg, 0.64 mmol, 5 mol%) and K3PO4 (6.7 g, 31.8 mmol, 2.5 equiv) were added. The mixture was refluxed for 2 h, followed by TLC. The crude residue was purified by column chromatography (c -hexane/ethyl acetate 4/1 to 1/1) to afford 3.0 g of 4 as a white solid (80% yield).
Chemical Formula: C18H13CIN2 ; Molecular Weight: 292.77 g/mol ; Melting Point: 159 °C
IR (KBr disc): 3045, 3019, 2924, 2853, 1638, 1578, 1446, 1427, 1385, 1166, 1087, 1057, 1023, 951, 921, 804, 729, 714, 687 cm-1.
!H-NMR (CDCb) d 8.90 (d, / = 2.4 Hz, 1H, HPyr), 8.72 (dd, / = 4.9, 1.6 Hz, 1H, HPyr), 8.51 (d, / = 2.4 Hz, 1H, HPyr), 8.16 (d, / = 2.4 Hz, 1H, HPyr), 7.95 (ddd, / = 7.9, 2.4, 1.6 Hz, 1H, HPyr), 7.61-7.59 (m, 2H, HAT), 7.49-7.41 (m, 4H, HPyr, 3 HAT, Ar-CH=C//-Pyr), 7.38-7.34 (m, 1H, HAT), 7.21 (d, / = 16.3 Hz, 1H, Ar-C//=CH-Pyr).
13C-NMR (CDCb) d 150.0, 149.5, 147.9, 146.1, 136.2, 136.1, 134.7, 134.0 (Ar-CH=CH- Pyr), 132.9, 132.9, 132.5, 132.3, 128.9 (2 CATH), 127.1 (2 CATH), 123.9, 122.7 (Ar-CH=CH- Pyr).
MS (ESI+): m/z 293.35 [M + H+], 295.34 [M+ + 2]
(/?)-6-Iodo-5-styryl-3,3'-bipyridine 5. To a solution of (C)-6-chloro-5-styryl-3,3'- bipyridine 4 (3.5 g, 12.1 mmol) in acetonitrile, Nal (4.5 g, 4.8 mmol, 2.5 equiv) and acetyl chloride (1.4 g, 18.1 mmol, 1.5 equiv) were added. The reaction mixture was refluxed for 16 h, affording 4.3 g of crude residue (80% of 5, by 1 H-N R) as a yellow solid which was used in the next reaction without further purification.
Chemical Formula: C18H13IN2 ; Molecular Weight: 384.22 g/mol ; Melting Point: 160 °C
IR (KBr disc): 3079, 3045, 3023, 2924, 1634, 1574, 1544, 1495, 1453, 1420, 1381, 1163, 1087, 1060, 1023, 959, 914, 805, 729, 712, 680 cm 1.2"
!H-NMR (CDCb) d 8.89 (dd, 7 = 2.4, 0.9 Hz, 1H, HPyr), 8.71 (dd, 7= 4.8, 1.7 Hz, 1H, HPyr), 8.46 (d, 7 = 2.4 Hz, 1H, HPyr), 7.95 (d, 7 = 2.4 Hz, 1H, HPyr), 7.93 (ddd, 7 = 7.9, 2.4, 1.7 Hz, 1H, HPyr), 7.61-7.59 (m, 2H, HAT), 7.48-7.41 (m, 3 H, 2HAT, Pyr-H), 7.38-7.34 (m, 2H, HAT), 7.29 (d, 7 = 16.1 Hz, 1H, Ar-CH=C//-Pyr), 7.11 (d, 7 = 16.1 Hz, 1H, Ar-C//=CH-Pyr). 13C-NMR (CDCb) d 149.1, 148.0, 144.8, 140.7, 135.4, 135.2, 134.1, 133.4, 133.2, 132.9, 128.7 (2 CATH), 128.0, 127.4, 126.4 (2 CATH), 124.0, 115.9.
MS (ESI+): m/z 385.41 [M + H+], 386.40 [M+ + 1]
(£')-6-Bromo-5-methyl-3'-styryl-3,2':5',3"-terpyridine 6. To a reaction vessel containing (£)-6-iodo-5-styryl-3, 3 '-bipyridine 5 (1.5 g, 3.9 mmol) in 1,4-dioxane under nitrogen atmosphere, (6-bromo-5-methylpyridin-3-yl)boronic acid (930 mg, 4.3 mmol, 1.1 equiv), tetrakis(triphenylphosphine)palladium(0) (225 mg, 0.19 mmol, 5 mol%) and K3PO4 (2.0 g , 9.8 mmol, 2.5 equiv) were added. The mixture was refluxed for 4 h, followed by TLC. The crude residue was purified by column chromatography (c-hexane/ethyl acetate 95/5 to 4/1) to afford 751 mg of 6 as a yellow solid (45% yield).
Chemical Formula: C24HisBrN3 ; Molecular Weight: 428.33 g/mol ; Melting Point: 149 °C
IR (KBr disc): 3039, 1588, 1557, 1490, 1438, 1379, 1190, 1120, 1053, 1023, 970, 907, 804, 746, 708, 721, 686, 540 cm-1.
3H-NMR (CDCb) d 8.96 (dd, 7 = 2.4, 0.8 Hz, 1H, HPyr), 8.83 (d, 7 = 2.2 Hz, 1H, HPyr), 8.72 (dd, 7= 4.8, 1.6 Hz, 1H, HPyr), 8.50 (d, 7= 2.4 Hz, 1H, HPyr), 8.21 (d, 7 = 2.2 Hz, 1H, HPyr), 8.00 (dt, 7 = 7.9, 2.0 Hz, 1H, HPyr), 7.92 (d, 7 = 2.4 Hz, 1H, HPyr), 7.50-7.44 (m, 3H, 2 HAT, HPyr), 7.40-7.29 (m, 3H, HAT), 7.22 (d, 7 = 16.2 Hz, 1H, Ar-CH=C//-Pyr), 7.14 (d, 7 = 16.2 Hz, 1H, Ar-Cf/=CH-Pyr), 2.49 (s, 3H, CH ).
13C-NMR (CDCb) d 152.8, 149.8, 148.4, 148.1, 147.0, 145.0, 139.9, 136.4, 135.3, 134.7, 134.6, 133.5, 133.2, 133.1, 132.8, 132.2, 129.0 (2 CATH), 128.8, 127.0 (2 CATH), 124.5, 124.0, 22.2 (CH ). MS (ESI+): m/z 428.24 [M + H+], 430.29 [M+ + 2]
(£')-5-Bromo-2-iodo-3-styrylpyridine 7. To a reaction vessel containing (£)-5-bromo-2- chloro-3-styrylpyridine 3 (4.8 g, 16.3 mmol) in acetonitrile, Nal (6.1 g, 40.8 mmol, 2.5 equiv) and acetyl chloride (2.0 g, 24.5 mmol, 1.5 equiv) were added. The reaction mixture was refluxed for 12 h, affording 5.22 g of crude residue (80% of 7, by 1 H-N R) as yellow solid which was used in the next reaction without further purification.
Chemical Formula: C13H9BHN ; Molecular Weight: 386.03 g/mol ; Melting Point: 78 °C IR (KBr disc): 3054, 3023, 1629, 1492, 1446, 1377, 1269, 1109, 1026, 952, 889, 751 cm-1. !H-NMR (CDCb) d 8.29 (d, / = 2.0 Hz, 1H, HPyr), 7.83 (d, / = 2.0 Hz, 1H, HPyr), 7.52 (d, / = 7.8 Hz, 2H, HAT), 7.38 (t, J = 7.8 Hz, 2H, HAT), 7.32 (d, J = 7.8 Hz, 1H, HAT), 7.09 (d, 1H, = 16.1 Hz, Ar-CH=C7/-Pyr), 6.94 (d, / = 16.1 Hz, 1H, Ar-C//=CH-Pyr).
13C-NMR (CDCb) d 149.6, 139.6, 135.9, 135.1, 134.7, 129.0, 128.9 (2 CATH), 128.1, 127.1 (2 CATH), 121.1, 120.8.
MS (ESI+): m/z 386.31 [M + H+], 388.24 [M+ + 2]
(/?)-5,6'-Dibromo-5'-methyl-3-styryl-2,3'-bipyridine 8. To a reaction vessel containing ( £) - 5 - b ro m o - 2 - i o do - 3 - s t y r y 1 p y r i d i n c 7 (3.0 g, 7.7 mmol) in 1,4-dioxane under nitrogen atmosphere, (6-bromo-5-methylpyridin-3-yl)boronic acid (1.8 g, 8.4 mmol, 1.1 equiv), tetrakis(triphenylphosphine)palladium(0) (445 mg, 0.38 mmol, 5 mol%) and K3PO4 (4.1 g , 19.3 mmol, 2.5 equiv) were added. The mixture was refluxed for 4 h, followed by TLC. The crude residue was purified by column chromatography (c-hexane/ethyl acetate 99/1 to 95/5) to afford 1.8 g of 8 as a yellow solid (55% yield). Chemical Formula: CigHuBnNi ; Molecular Weight: 430.14 g/mol ; Melting Point: 143 °C
IR (KBr disc): 3031, 1393, 1111, 1050, 968, 898, 740, 685, 499 cm-1.
!H-NMR (CDCb) d 8.65 (d, / = 1.9 Hz, 1H, HPyr), 8,41 (d, / = 1.9 Hz, 1H, HPyr), 8.17 (d, J = 1.9 Hz, 1H, HPyr), 7.83 (d, /= 1.9 Hz, 1H, HPyr), 7.42 (d, 7 = 7.8 Hz, 2H, HAT), 7.36 (t, / = 7.8 Hz, 2H, HAT), 7.31 (d, / = 7.8 Hz, 1H, HAT), 7.12 (d, /= 16.6 Hz, 1H, Ar-CH=C//-Pyr), 6,99 (d, /= 16.6 Hz, 1H, Ar-C//=CH-Pyr), 2.46 (s, 3H, CH ).
13C-NMR (CDCb) d 151.4, 149.4, 147.8, 144.9, 139.6, 136.5, 136.0, 135.2, 134.0, 133.9, 133.3, 128.9 (2 CATH 128.8, 126.9 (2 CATH), 123.4, 120.4, 22.0 (CH3).
MS (ESI+): m/z 431.51 [M + H+], 433.53 [M+ + 2]
(/?)-5-Bromo-6'-iodo-5'-methyl-3-styryl-2,3'-bipyridine 9. To a solution of (£)- 5,6'- dibromo-5'-methyl-3-styryl-2,3'-bipyridine 8 (3.8 g, 8.9 mmol) in acetonitrile, Nal (3.3 g, 22.3 mmol, 2.5 equiv) and acetyl chloride (1.0 g, 13.4 mmol, 1.5 equiv) were added. The reaction mixture was refluxed for 8 h, affording 4.0 g of crude residue (90% of 9, by CH- NMR) as yellow solid which was used in the next reaction without further purification. Chemical Formula: Ci9Hi4BrIN2 ; Molecular Weight: 477.14 g/mol ; Melting Point: 147 °C
IR (KBr disc): 3029, 1576, 1449, 1428, 1389, 1112, 1089, 968, 900, 686, 498 cm-1. !H-NMR (CDCb) d 8.66 (d, /= 2.3 Hz, 1H, HPyr), 8.38 (d, / = 2.3 Hz, 1H, HPyr), 8.16 (d, J = 2.3 Hz, 1H, HPyr), 7.73 (d, / = 2.3 Hz, 1H, HPyr), 7.44 (d, /= 7.4 Hz, 2H, HAT), 7.38 (t, / = 7.4 Hz, 2H, HAT), 7.34-7.30 (m, 1H, HAT), 7.14 (d, / = 16.2 Hz, 1H, Ar-CH=C//-Pyr), 6.98 (d, /= 16.2 Hz, 1H, Ar-C7/=CH-Pyr), 2.48 (s, 3H, CH ).
13C-NMR (CDCb) d 151.4, 149.5, 148.1, 139.2, 137.5, 136.5, 135.9, 134.0, 133.9, 133.3, 128.9 (2 CATH), 128.8, 126.9 (2 CATH), 125.6, 123.4, 120.4, 26.2 (CH ).
MS (ESI+): m/z 477.48 [M + H+], 479.54 [M+ + 2]
(/?)-5-Bromo-5'-methyl-3-styryl-2,3':6',3"-terpyridine 10. To a reaction vessel containing (£,)-5-bromo-6'-iodo-5'-methyl-3-styryl-2,3'-bipyridine 9 (2.0 g, 4.2 mmol) in 1,4-dioxane under nitrogen atmosphere, pyridin-3-ylboronic acid (570 mg, 4.6 mmol, 1.1 equiv), tetrakis(triphenylphosphine)palladium(0) (243 mg, 0.2 mmol, 5 mol%) and K3P04 (2.3 g , 10.5 mmol, 2.5 equiv) were added. The mixture was refluxed for 3 h, followed by TLC. The crude residue was purified by column chromatography (c -hexane/ethyl acetate 8/2 to 1/1) to afford 1.1 g of 10 as an off-white solid (60% yield).
Chemical Formula: C24HisBrN3 ; Molecular Weight: 428.33 g/mol ; Melting Point: 136- 138 °C
IR (KBr disc): 3032, 2917, 1635, 1414, 1384, 1118, 950, 740, 541 cm-1.
3H-NMR (CDCb) d 8.88 (dd, 7 = 2.3, 0.9 Hz, 1H, HPyr), 8.78 (dd, 7 = 2.1, 0.7 Hz, 1H, HPyr), 8.69 (d, 7 = 2.2 Hz, 1H, HPyr), 8.68 (dd, 7 = 4.9, 1.7 Hz, 1H, HPyr), 8.21 (d, 7 = 2.1 Hz, 1H, HPyr), 7.97 (ddd, 7 = 7.8, 2.3, 1.7 Hz, 1H, HPyr), 7.95 (dd, 7 = 2.2, 0.8 Hz, 1H, HPyr), 7.45 (m,
3H, 2 HAT, Hpyr), 7.38-7.28 (m, 3H, HAT), 7.16 (d, J = 16.2 Hz, 1H, Ar-CH=C//-Pyr), 7.11 (d, J = 16.2 Hz, 1H, Ar-C7/=CH-Pyr), 2.48 (s, 3H, CH ).
13C-NMR (CDCb) d 155.5, 152.4, 149.9, 149.6, 149.3, 148.1, 139.9, 136.9, 136.6, 136.3, 133.9, 133.8 (Ar-CH=CH-Pyr), 133.6, 132.3, 131.4, 129.0 (2 CATH), 128.9, 127.1 (2 CATH), 123.8 (Ar-CH=CH-Pyr), 123.5, 120.4, 20.2 (CH3).
MS (ESI+): m/z 428.74 [M + H+], 430.62 [M+ + 2]
HRMS (ESI): m/z Calcd. for [M+H]+: 428.0762 ; Found: 428.0772.
(/?)-5-bromo-6'-(2-hydroxyphenyl)-5'-methyl-3-styryl-2,3'-bipyridine 11. To a reaction vessel containing (£,)-5-bromo-6'-iodo-5'-methyl-3-styryl-2,3'-bipyridine 9 (356 mg, 0.75 mmol, 1.0 equiv.) in 1,4-dioxane (20 mL) under nitrogen atmosphere, 2- hydroxyphenylboronic acid (120 mg, 0.83 mmol, 1.1 equiv.), tetrakis(triphenylphosphine)palladium(0) (43 mg, 0.04 mmol, 5 mol%) and K3PO4 (398 mg, 1.88 mmol, 2.5 equiv.) were added. The mixture was refluxed for 24 h, followed by TLC. The crude residue was purified by column chromatography (cyclohexane/ethyl acetate 100/0 to 90/10) to afford 183 mg of 11 as an off-white solid (55% yield).
Chemical formula: C2sHi9BrN20 ; Molecular weight: 443.34 g/mol
NMR *H (400 MHz, CDCb): <5 (ppm) = 12.15 (s, 1H), 8.61 (dd, 7 = 9.6, 2.2 Hz, 2H), 8.13
(d, 7 = 2.2 Hz, 1H), 7.96 (d, 7 = 2.2 Hz, 1H), 7.56 (dd, 7 = 7.9, 1.7 Hz, 1H), 7.38 (d, 7 = 6.9
Hz, 1H), 7.29 (t, 7 = 7.4 Hz, 1H), 7.25 (d, 7 = 7.0 Hz, 1H), 7.09 (d, 7 = 16.2 Hz, 1H), 7.05
6.97 (m, 4H), 6.88 (td, 7 = 7.6, 1.3 Hz, 1H), 2.59 (s, 3H). NMR 13C (75 MHz, CDCb): <5 (ppm) = 157.98, 156.42, 151.84, 149.55, 144.98, 141.95, 136.62, 136.10, 133.78, 133.43, 132.61, 131.61, 130.89, 129.80, 128.99, 128.84, 126.93, 123.59, 121.24, 120.36, 118.56, 118.17, 21.94.
(/?)-5-bromo-6'-(2-propoxyphenyl)-5'-methyl-3-styryl-2,3'-bipyridine 12. To a reaction vessel containing (£,)-5-bromo-6'-(2-hydroxyphenyl)-5'-methyl-3-styryl-2,3'-bipyridine 11 (163 mg, 0.37 mmol, 1.0 equiv.) in DMF (10 mL), 1-iodopropane (54 pL, 0.55 mmol, 1.5 equiv.) and K2CO3 (102 mg, 0.74 mmol, 2 equiv.) were added. The reaction mixture was stirred at 50°C for 17 h. The mixture was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure to give the crude residue which was purified by column chromatography (cyclohexane/ethyl acetate 100/0 to 90/10) to afford 147 mg of 12 as a yellow solid (82% yield).
Chemical formula: C2SH25B1N2O ; Molecular weight: 485.43 g/mol NMR 3H (400 MHz, CDCb): <5 (ppm) = 8.65 (d, / = 2.1 Hz, 1H), 8.62 (d, J = 2.2 Hz, 1H), 8.14 (d, / = 2.2 Hz, 1H), 7.77 (dd, / = 2.2, 0.9 Hz, 1H), 7.39 - 7.35 (m, 2H), 7.34 - 7.21 (m, 5H), 7.07 (s, 2H), 7.00 (td, / = 7.5, 1.0 Hz, 1H), 6.91 (dd, / = 8.2, 1.0 Hz, 1H), 3.86 (t, / = 6.5 Hz, 2H), 2.19 (s, 1H), 1.61 (h, /= 7.3 Hz, 2H), 0.81 (t, /= 7.4 Hz, 3H).
NMR 13C (75 MHz, CDCb): <5 (ppm) = 157.47, 156.16, 149.35, 147.06, 138.04, 136.33, 136.29, 133.37, 133.19, 132.86, 132.74, 130.59, 129.86, 129.66, 128.84, 128.60, 126.91, 124.10, 120.76, 112.06, 69.86, 22.59, 19.10, 10.59.
( E )- 6' -(2-propoxyphenyl)-5 ' -Methyl-5-(6-fluoropyridin-3-yl)-3-styryl-2,3 ' : 6' ,3 " - terpyridine 13. To a reaction vessel containing (£,)-5-bromo-6'-(2-propoxyphenyl)-5'- methyl-3-styryl-2,3'-bipyridine 12 (147 mg, 0.303 mmol) in 1,4-dioxane under nitrogen atmosphere, commercial 6-fluoropyridin-3-ylboronic acid (47 mg, 0.333 mmol, 1.1 equiv), tetrakis(triphenylphosphine)palladium(0) (35 mg, 0.03 mmol, 10 mol%) and K3PO4 (225 mg, 1.06 mmol, 3.5 equiv) were added. The mixture was refluxed for 26 h, followed by TLC. The crude residue was purified by column chromatography (dichloromethane/ethyl acetate 100/0 to 50/50) to afford 17 mg of 13 as an yellow oil (13% yield).
Chemical Formula: C33H28FN3O Molecular Weight: 501.61 g/mol !H-NMR (CDCb) : 8.76 (d, 7 = 2.2 Hz, 1H), 8.73 (dd, 7 = 2.1, 0.7 Hz, 1H), 8.50 (dt, 7 = 2.7, 0.9 Hz, 1H), 8.13 (d, 7 = 2.3 Hz, 1H), 8.04 (ddd, 7 = 8.5, 7.5, 2.7 Hz, 1H), 7.85 (dd, 7 = 2.2, 0.8 Hz, 1H), 7.44 - 7.35 (m, 2H), 7.22 (s, 6H), 7.14 (d, 7 = 16.2 Hz, 1H), 7.06 (ddd, 7 = 8.4, 3.0, 0.7 Hz, 1H), 7.01 (td, 7 = 7.4, 1.0 Hz, 1H), 6.92 (d, 7= 8.1 Hz, 1H), 3.88 (t, 7= 6.5 Hz, 2H), 2.21 (s, 3H), 1.63 (dtd, 7= 13.9, 7.4, 6.5 Hz, 2H), 0.83 (t, 7= 7.4 Hz, 3H). 13C-NMR (CDCb) 163.7 (d, 7 = 240.9 Hz), 157.5, 156.2, 154.4, 147.2, 146.6, 146.1 (d, 7= 15.1 Hz), 139.9 (d, 7 = 8.1 Hz), 138.2, 136.5, 133.2, 132.8, 132.4, 131.9, 131.6, 131.4 (d, 7 = 4.7 Hz), 130.6, 129.9, 129.7, 128.9, 128.5, 126.9, 124.9, 120.8, 112.1, 110.1 (d, 7 = 37.5 Hz), 69.9, 22.6, 19.1, 10.6.
19F NMR (376 MHz, CDCb) d (ppm) -68.49.
The final compound has been obtained from compound 12 as follows:
(/?)-5'-Methyl-5-(6-fluoropyridin-3-yl)-3-styryl-2,3':6',3"-terpyridine 14. To a reaction vessel containing (£')-5-bromo-5'-methyl-3-styryl-2,3':6',3"-terpyridine 10 (214 mg, 0.5 mmol) in 1,4-dioxane under nitrogen atmosphere, commercial 6-fluoropyridin-3-ylboronic acid (82 mg, 0.55 mmol, 1.1 equiv), tetrakis(triphenylphosphine)palladium(0) (58 mg, 0.05 mmol, 10 mol%) and K3PO4 (371 mg, 1.75 mmol, 3.5 equiv) were added. The mixture was refluxed for 12 h, followed by TLC. The crude residue was purified by column chromatography (dichloromethane/ethyl acetate 100/0 to 20/80) to afford 113 mg of 14 as an off-white solid (51% yield).
Chemical Formula: C29H21FN4 ; Molecular Weight: 444.51 g/mol ; Melting Point: 212 °C
NMR *H (400 MHz, CDCb): <5 (ppm) = 8.85 - 8.83 (m, 1H), 8.79 (d, 7 = 2.1 Hz, 1H), 8.77 (d, 7 = 2.2 Hz, 1H), 8.62 (dd, 7 = 4.9, 1.7 Hz, 1H), 8.53 - 8.48 (m, 1H), 8.13 (d, 7 = 2.2 Hz, 1H), 8.05 (ddd, 7= 8.5, 7.5, 2.6 Hz, 1H), 7.96 (dd, 7= 2.2, 0.8 Hz, 1H), 7.92 (dt, 7= 7.9, 1.9 Hz, 1H), 7.44 - 7.35 (m, 3H), 7.33 - 7.27 (m, 2H), 7.27 - 7.21 (m, 1H), 7.18 (d, 7 = 2.2 Hz,
1H), 7.06 (ddd, 7= 8.5, 3.1, 0.7 Hz, 1H), 2.44 (s, 3H).
NMR 13C (75 MHz, CDCb): <5 (ppm) = 163.75 (d, 7= 241.2 Hz), 155.41, 153.59, 149.92, 149.26, 148.10, 146.76, 146.14 (d, 7 = 15.1 Hz), 139.99 - 139.75 (m), 136.53 (d, 7 = 35.2 Hz), 134.09, 133.32, 132.59, 132.15, 131.90, 131.29, 131.23, 128.92, 128.67, 126.91, 124.60, 110.13 (d, 7 = 37.5 Hz), 20.07.
NMR 19F (376 MHz, CDCb): <5 (ppm) = - 68.28
(/?)-5'-methyl-5-(6-(amino-propan-l-ol)pyridin-3-yl)-3-styryl-2,3':6',3"-terpyridine
15. The 3-aminopropan-l-ol (19 pL, 0.24 mmol, 2 equiv.) was dissolved in dry DMF and DIPEA (N,N-diisopropylethylamine) (52 pL, 0.3 mmol, 2.5 equiv). (/?)-5'-methyl-5-(6- fluoropyridin-3-yl)-3-styryl-2,3':6',3"-terpyridine 14 (53 mg, 0.12 mmol, 1.0 equiv) was added, and the mixture was stirred at 120 °C under microwave irradiation for 5 h. After cooling to rt, the mixture was diluted with EtOAc and washed with water and brine. The organic layers were dried over MgSCb, filtered and concentrated in vacuo. The crude residue was purified by column chromatography (EtOAc/MeOH 100/0 to 90/10) to afford 36 mg of 15 as an pale yellow solid (61% yield).
Aspect: yellow powder
Chemical Formula: C32H29N5O; Molecular Weight: 499.62 g/mol NMR *H (400 MHz, CDCb): <5 (ppm) = 8.83 (d, 7 = 2.3 Hz, 1H), 8.77 (d, 7 = 2.1 Hz, 1H), 8.72 (d, 7 = 2.2 Hz, 1H), 8.60 (dd, 7 = 4.9, 1.7 Hz, 1H), 8.36 (d, 7 = 2.5 Hz, 1H), 8.07 (d, 7 = 2.3 Hz, 1H), 7.94 (d, 7 = 2.1 Hz, 1H), 7.91 (dt, 7 = 7.9, 2.1 Hz, 1H), 7.67 (dd, 7 = 8.6, 2.5 Hz, 1H), 7.44 - 7.33 (m, 3H), 7.32 - 7.26 (m, 2H), 7.25 - 7.21 (m, 1H), 7.18 (d, / = 15.9 Hz, 1H), 7.15 (d, / = 16.5 Hz, 1H), 6.49 (d, / = 8.6 Hz, 1H), 4.87 (t, / = 6.4 Hz, 1H), 3.64 (t, / = 5.5 Hz, 2H), 3.61 - 3.50 (m, 2H), 2.42 (s, 3H), 1.75 (p, / = 5.8 Hz, 2H).
NMR 13C (75 MHz, CDCb): <5 (ppm) = 158.94, 150.00, 149.24, 148.14, 146.20, 146.07, 139.85, 136.69, 136.60, 136.06, 133.42, 132.71, 131.22, 131.17, 128.86, 128.43, 126.86, 125.05, 123.28, 108.72, 58.84, 38.09, 33.34, 20.05.
2-(2,6-dioxopiperidin-3-yl)-4-fluoro-2,3-dihydro-///-isoindole-l,3-dione 16. In a round- bottom flask, commercial 3-fluorophthalic anhydride (171 mg, 1 mmol), commercial 3- aminopiperidine-2,6-dione hydrochloride (169 mg, 1 mmol), and sodium acetate (121 mg, 1.2 mmol) were mixed in AcOH (30 mL). The resulting reaction mixture was heated to reflux at 90°C for 16 h. After cooling to room temperature, most of the AcOH was evaporated and the purple residue was dissolved in water (20 mL) then extracted with ethyl acetate (3 x 50 mL). The combined organic layer was dried (brine then MgS04) then concentrated under vacuo. The residue is purified by flash column chromatography with cyclohexane/ethyl acetate (50/50) to obtain the product as a pale blue solid (Yield: 68 %).
Chemical formula: C13H9FN2O4 ; Molecular weight: 276.22 g/mol NMR 3H (400 MHz, DMSO-d6): <5 (ppm) = 11.15 (s, 1H, NH), 7.99 - 7.92 (m, 1H, ¾), 7.80 (d, 3J = 7.2 Hz, 1H, H7), 7.77-7.72 (m, 1H, H5), 5.16 (dd, 3Jax-ax= 12.8, 3Jax-eq = 5.4 Hz, 1H, H3 , 2.96 - 2.84 (m, 1H, ¾·), 2.65 - 2.52 (m, 2H, H4 , ¾·), 2.11 - 2.02 (m, 1H, ¾·) NMR 13C (75 MHz, DMSO-d6): <5 (ppm) = 173.2 (C6’ 170.2 (C2 , 166.6 (d, 3JCF = 2.9 Hz, C3), 164.4 (Ci), 157.3 (d, 1JCF = 262.4 Hz, C4), 138.5 (d, 3JCF = 7.9 Hz, C6), 133.9 (d, 3JCF = 1.2 Hz, C7a), 123.5 (d, 2JCF = 19.6 Hz, Cs), 120.5 (d, 4JCF = 3.2 Hz, C7), 117.5 (d, 2JCF = 12.7 Hz, C3a), 49.5 (C , 31.4 (Gr), 22.3 (Cs·)
NMR 19F ( MHz, DMSO-d6): <5 (ppm) = -114.7
MS (ESI+): m/z: 308.90 [M+MeOH+H]+, 638.52 [M+2MeCN+2H]2+, 552.59 [2M+H]+
2-(2,6-dioxopiperidin-3-yl)-4-((2-(2-(prop-2-yn-l-yloxy)ethoxy)ethyl)amino) isoindoline-l,3-dione 17. In a round-bottom flask, 4-fluoro-thalidomide 16 (120 mg, 0.44 mmol), amine linker (75 pL, 0.53 mmol, 1.2 eq.), and DIEA (150 pL, 0.87 mmol, 3 eq.) in dry DMF (5 mL) were stirred at 90°C for 16 h. The reaction mixture was diluted with 20 mL of ethyl acetate and the organic phase was washed with water (5 x 10 mL) then dried (brine, then MgS04) and evaporated under reduce pressure. The residue was purified by column chromatography using a gradient of ethyl acetate in cyclohexane (0% to 70%) to afford the product as a green solid (Yield: 32 %).
Chemical formula: C20H21N3O6 ; Molecular weight: 399.40 g/mol NMR ¾ (400 MHz, DMSCM6): <5 (ppm) = 11.16 (s, 1H, NHa), 7.64 (dd, 3J = 8.5, 3J = 12 Hz, 1H, H6), 7.20 (d, J = 8.6 Hz, 1H, H5), 7.09 (d, J = 7.2 Hz, 1H, H7), 6.66 (t, 3J = 5.6 Hz, 1H, NHb), 5.11 (dd, 3Jax-ax= 12.9, 3Jax-eq= 5.5 Hz, 1H, ¾·), 4.19 (d, 4J = 2.4 Hz, 2H, ¾··), 3.69 - 3.65 (m, 2H, ¾··), 3.65 - 3.60 (m, 4H, H3 , ¾··), 3.53 (q, 3J= 5.6 Hz, 2H, Hi”), 3.46 (t, 4J= 2.4 Hz, 1H, Hr·), 2.99 - 2.88 (m, 1H, ¾·), 2.69 - 2.58 (m, 2H, H4\ Hs ), 2.13 - 2.04 (m, 1H, HsO.
NMR 13C (75 MHz, DMSCM6): <5 (ppm) = 172.9 (C6·), 170.2 (C2 , 169.0 (C3), 167.3 (Ci), 146.4 (C4), 136.3 (C6), 132.1 (C7a), 117.5 (Cs), 110.7 (C7), 109.3 (C3a), 80.4 (C6”X 77.2
(CrO, 69.5 (C3··), 68.9 (C2”X 68.6 (C4·), 57.6 (Cs··), 48.6 (C3·), 41.7 (CrO, 31.0 (Cr), 22.2 (CsO
18 30% 19 90% 20 51%
Methyl 3-bromo-4-hydroxybenzoate 18. To a solution of methyl 4-hydroxybenzoate (1 g, 6.57 mmol, 1.0 equiv.) in dry CH2CI2 (75 mL) at room temperature was added carefully dibromide (0.33 mL, 65.7 mmol, 10.0 equiv.). The mixture was stirred at room temperature for 18 h. Then acetic acid (1 mL) was added and the reaction was stirred for 3 h more. The mixture was washed with saturated aqueous NaiSiCL, brine, dried (MgS04) and concentrated under vaccuo. The resulting crude residue was purified by column chromatography (C!LCh/McOH 100/0 to 99/1) to afford 592 mg of 18 as a white solid (39%).
Chemical Formula: CsH7Br03; Molecular Weight: 231.05 g/mol ; Melting Point: 109°C NMR ¾ (400 MHz, CDCb): <5 (ppm) = 8.12 (d, / = 2.1 Hz, 1H), 7.85 (dd, /= 8.5, 2.0 Hz, 1H), 6.98 (d, / = 8.5 Hz, 1H), 5.90 (s, 1H), 3.83 (s, 3H). NMR 13C (75 MHz, CDCb): <5 (ppm) = 165.6, 156.2, 134.0, 131.1, 124.1, 115.8, 110.1, 52.2.
Methyl 3-bromo-4-propoxybenzoate 19. To a solution of methyl 3-bromo-4- hydroxybenzoate 18 (462 mg, 2 mmol, 1.0 equiv.) in dry acetone (20 mL) at room temperature were added 1-iodopropane (0.29 mL, 3 mmol, 1.5 equiv.) and K2CO3 (553 mg, 4 mmol, 2 equiv.). After cooling to room temperature, the mixture was diluted with EtOAc and washed with water, brine, dried (MgSCb) and concentrated under vaccuo to afford 492 mg of 19 as a pale yellow oil (90%).
Chemical Formula: CnHisBrCb; Molecular Weight: 273.13 g/mol NMR 3H (400 MHz, CDCb): <5 (ppm) = 8.14 (d, J = 2.1 Hz, 1H), 7.86 (dd, J = 8.6, 2.1 Hz, 1H), 6.79 (d, J = 8.7 Hz, 1H), 3.96 (t, J = 6.4 Hz, 2H), 3.81 (s, 3H), 1.80 (h, J = 7.3 Hz, 3H), 1.01 (t, J = 7.4 Hz, 3H).
NMR 13C (75 MHz, CDCb): <5 (ppm) = 165.80, 159.09, 134.76, 130.51, 123.38, 111.83, 111.81, 70.76, 52.11, 22.38, 10.51
Methyl 3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-4-(propoxy) benzoate 20. To a reaction vessel containing Methyl 3-bromo-4-propoxybenzoate 19 (492 mg, 1.74 mmol,
1.0 equiv.) in 1,4-dioxane (3.5 mL) under nitrogen atmosphere, commercial bis(pinacolato)diboron (664 mg, 2.62 mmol, 1.5 equiv.), bis(triphenylphosphine)palladium(II) dichloride (250 mg, 0.35 mmol, 2 mol%) and KOAc (514 mg, 5.23 mmol, 3.0 equiv.) were added. The mixture was refluxed for 24 h, followed by TLC. The crude residue was purified by column chromatography (cyclohexane/ethyl acetate 100/0 to 80/20) to afford 285 mg of 20 as a white solid (51% yield).
Chemical Formula: C17H25BO5; Molecular Weight: 320.19 g/mol ; Melting Point: 87°C NMR 3H (400 MHz, CDCb): d (ppm) = 8.22 (d, /= 2.4 Hz, 1H), 7.99 (dd, / = 8.7, 2.4 Hz, 1H), 6.77 (d, / = 8.8 Hz, 1H), 3.91 (t, / = 6.2 Hz, 2H), 3.80 (s, 3H), 1.83 - 1.71 (m, 2H), 1.19 (s, 12H), 1.01 (t, /= 7.4 Hz, 3H).
NMR 13C (75 MHz, CDCb): d (ppm) = 167.2, 166.9, 138.1, 134.4, 121.8, 110.6, 83.5, 69.8, 51.8, 25.0, 22.6, 10.5. nB NMR (128 MHz, CDCb) d (ppm): 30.73.
(/?)-5'-Methyl-5-(methyl 4-(propoxy)benzoate)-3-styryl-2,3':6',3"-terpyridine 21. To a reaction vessel containing (£')-5-Bromo-5'-methyl-3-styryl-2,3':6',3"-terpyridine 10 (209 mg, 0.49 mmol, 1.0 equiv.) in 1,4-dioxane (20 mL) under nitrogen atmosphere, methyl 3- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-4-(propoxy) benzoate 20 (235 mg, 0.73 mmol, 1.5 equiv.), tetrakis(triphenylphosphine)palladium(0) (5.48 mg, 0.025 mmol, 5 mol%) and K3PO4 (363 mg, 1.71 mmol, 3.5 equiv.) were added. The mixture was refluxed for 19 h, followed by TLC. The crude residue was purified by column chromatography (0 hexane/ethyl acetate 8/2 to 1/1) to afford 45 mg of 21 as a white solid (17% yield). Chemical Formula: C35H31N3O3 ; Molecular Weight: 541.24 g/mol NMR *H (400 MHz, CDCb): <5 (ppm) = 8.85 - 8.83 (m, 1H), 8.81 - 8.77 (m, 2H), 8.64 - 8.59 (m, 1H), 8.20 (d, / = 2.1 Hz, 1H), 8.09 (d, / = 2.2 Hz, 1H), 8.04 (dd, / = 8.7, 2.2 Hz,
1H), 7.97 (dd, / = 2.2, 0.8 Hz, 1H), 7.92 (dt, / = 7.8, 1.9 Hz, 1H), 7.42 - 7.35 (m, 3H), 7.32 - 7.26 (m, 2H), 7.25 - 7.22 (m, 1H), 7.20 (d, / = 15.8 Hz, 1H), 7.12 (d, / = 16.2 Hz, 1H), 7.00 (d, / = 8.6 Hz, 1H), 4.03 (t, / = 6.4 Hz, 2H), 3.87 (s, 3H), 2.43 (s, 3H), 1.87 - 1.71 (m,
2H), 0.99 (t, 7 = 7.4 Hz, 3H). dry DMF HCI 1 M 60°C, 48h Et20/EtOAc (1:1) rt, ovn n = 1,2 22a (n=1) 55% 23a (n=1) 52% 22b (n=2) 77% 23b (n=2) 54%
Nucleophilic substitution:
1,3-diazidopropane 21a and 1,4-diazidobutane 22b. Alkyl diazides were obtained by a trivial nucleophilic displacement reaction between the proper alkyl dihalide and sozium azide, according to the following procedure. The proper dibromoalkyle (10.0 mmol, namely 2.02 g of 1,3-dibromopropane) was dissolved in dry DMF (10 mL) under nitrogen and NaN3 (1.30 g, 20.0 mmol) was added. The mixture was kept under magnetic stirring at 60°C for 24 h. The reaction mixture was then poured into water (50 mL) and extracted five times with diethyl ether (50 mL each). The organic extracts were washed with water (5 mL) then dried (brine, then MgS04). The reaction mixture was evaporated under reduced pressure to yield crude oily products.
22a. Chemical formula: C3H6N6 ; Molecular weight: 126.12 g/mol ; Aspect: colorless oil Yield: 55 %
NMR 3H (400 MHz, DMSO-rf6): <5 (ppm) = 3.42 (t, 3J = 6.7 Hz, 4H, Hi, HO, 1.77 (p, 3J = 6.7 Hz, 2H, H2)
NMR 13C (75 MHz, DMSO-rf6): <5 (ppm) = 48.5 (Ci, C3), 28.1 (C2)
22b. Chemical formula: C4H8N6 ; Molecular weight: 140.15 g/mol ; Aspect: colourless oil
Yield: 77 %
NMR 3H (400 MHz, DMSO-rf6): <5 (ppm) = 3.40 - 3.28 (m, 4H, Hi, H4), 1.76 - 1.66 (m, 4H, H2, ¾)
NMR 13C (75 MHz, DMSO-d6): <5 (ppm) = 50.9 (Ci, C4), 26.2 (C2, C3)
3-azidopropylamine 23a and 4-azidobutylamine 23b. Azidoalkylamines were obtained by a regioselective Staudinger reduction of diazide compounds 22. To a mixture of diazidoalkyl compound 22 (10 mmol) cooled to 0°C, aqueous 1M HC1 (20 ruL), diethyl ether (7 ruL), and ethyl acetate (7 ruL) was added. Triphenylphosphine was added portionwise during 30 min. The mixture was warmed to rt and stirred for an additional 20 h, and then the organic layer was separated from the aqueous layer. The aqueous phase was washed with ethyl ether (2 x 50 ruL) to remove triphenylphosphine oxide residual. The aqueous phase was basified to a pH 13 by aqueous NaOH and then was extracted with dichloromethane (3 * 100 ruL). The combined dichloromethane layer was dried over MgS04 and concentrated to yield 4-azidoalkylamine derivative as an oil.
23a. Chemical formula: C4HxN4 ; Molecular weight: 100.12 g/mol ; Aspect: yellow oil Yield: 52 %
NMR 3H (400 MHz, DMSCM6): <5 (ppm) = 3.38 (t, 3J = 6.7 Hz, 2H, Hi), 2.59 (t, 3J = 6.7 Hz, 2H, ¾), 1.59 (p, 3J= 6.7 Hz, 2H, H2)
NMR 13C (75 MHz, DMSO-rf6): <5 (ppm) = 48.9 (C3), 39.2 (Ci), 32.6 (C2)
23b. Chemical formula: C4HION4 Molecular weight: 114.15 g/mol ; Aspect: yellow oil Yield: 54 %
NMR 3H (400 MHz, DMSO-rf6): <5 (ppm) = 3.23 (t, 3/ = 6.8 Hz, 2H, Hi), 2.67 (t, 3/ = 6.9 Hz, 2H, HO, 1.65 - 1.53 (m, 2H, H2), 1.51 - 1.44 (m, 2H, H4). NMR 13C (75 MHz, DMSC 6): <5 (ppm) = 51.4 (C4), 41.7 (Ci), 30.8 (C2), 26.3
(C3).
Synthesis of one of the preferred compounds: CRBN-PBMl-001
CRBN-PBMl-001 was obtained by a Sonogashira cross -coupling reaction. Under inert atmosphere, triethylamine (42 pL, 0.3 mmol, 3.0 equiv.), copper (I) iodide (2 mg, 0.01 mmol, 10 %mol), 2-(2,6-dioxopiperidin-3-yl)-4-((2-(2-(prop-2-yn-l-yloxy) ethoxy)ethyl)amino)isoindoline-l,3-dione 17 (44 mg, 0.11 mmol, 1.1 equiv) and tetrakis(triphenylphosphine)palladium(0) (8 mg, 0.005 mmol, 5 %mol) were added to a solution of 10 (42.8 mg, 0.10 mmol, 1.0 equiv) in dry DMF (2 mL). The reaction mixture was stirred at 50°C for 6h. The mixture was diluted with ethyl acetate. The aqueous layers were washed with 4 mL of a saturated solution of NaHCCL and with brine. The combined organic layers were dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The crude residue was purified by flash chromatography on silica gel column (water/acetonitrile 100/0 to 0/100) to afford 41 mg of CRBN-PBMl-001 as a solid yellow (55 % yield).
Chemical formula: C44H38N6O6 ; Molecular weight: 746.82 g/mol NMR 3H (400 MHz, CDCb): <5 (ppm) = 8.82 (s, 1H), 8.75 - 8.66 (m, 2H), 8.64 - 8.58 (m, 3H), 8.14 (d, J = 2.2 Hz, 1H), 8.03 (d, J = 1.9 Hz, 1H), 7.94 - 7.84 (m, 3H), 7.69 - 7.60 (m, 1H), 7.43 - 7.33 (m, 4H), 7.31 - 7.20 (m, 3H), 7.09 - 6.99 (m, 3H), 6.85 (d, J = 8.5 Hz, 1H), 6.45 (t, J = 5.5 Hz, 1H), 4.83 (dd, J = 12.1, 5.4 Hz, 1H), 4.44 (s, 1H), 3.81 - 3.64 (m, 4H), 3.43 (q, J = 5.5 Hz, 1H), 2.82 - 2.57 (m, 2H), 2.40 (s, 3H), 2.06 - 1.97 (m, 1H).
NMR 13C (75 MHz, CDCb): <5 (ppm) = 171.2, 169.3, 168.5, 167.6, 155.4, 150.8, 149.9, 149.9, 149.5, 149.3, 149.3, 148.0, 147.9, 146.8, 139.9, 139.8, 136.9, 136.69, 136.65, 136.5, 136.4, 136.2, 136.1, 135.8, 133.9, 133.7, 133.3, 132.5, 131.4, 131.3, 128.9, 128.9, 128.8, 128.6, 126.9, 126.9, 124.2, 123.7, 123.3, 119.2, 116.8, 111.7, 89.3, 82.9, 70.6, 69.6, 69.5, 59.3, 48.9, 42.4, 31.5, 22.8, 20.0, 20.0.
MS (ESI+): m/z: 373.93 [M+2H]2+, 748.36 [M+H]+ Example 4: Further biological activity of compounds of the invention
Material and methods
Cell culture and drug testing
Human platinum resistant ovarian carcinoma cells IGROV1-R10 were used. This cell line was grown in RPMI 1640 medium supplemented with 2 mM GlutamaxTM, 25 mM HEPES, 10% fetal calf serum, and 33 mM sodium bicarbonate (Fisher Scientific, Illkirch, France). Cells were maintained at 37°C in a humid atmosphere with 5% C02. Cells were seeded in 25 cm2 flasks. After 24h, exponentially growing cells were exposed to 1 mM of compounds for 24h. CRBN-PBMl-001 targeting both Mcl-1 and CRBN and Pyridoclax were designed and synthetized by the CERMN (Centre d’Etudes et de Recherche sur le Medicament de Normandie) and dissolved in DiMethyl SulfOxide (DMSO, Sigma- Aldrich).
Target expression analysis by western blot
Cells were rinsed with ice-cold PBS, suspended in a lysis buffer [RIPA : NaCl 150 mM, Tris (pH 8) 50 mM, Triton X100 1%, PMSF 4 mM, EDTA 5 mM, NaF 10 mM, NaPPi 10 mM, Na3V04 1 mM, aprotinin 0.5 pL/mL and 4.6 mL ultra-pure water] and incubated on ice for 30 min. Lysates were collected after centrifugation (13200 g, 10 min, 4°C) and protein concentrations were determined using the Bradford assay (Bio-Rad, Hercules, USA). Equal amounts of protein (25 pg) were separated by SDS-PAGE on a 4-15% gradient polyacrylamide Mini-PROTEAN®TGX™ precast gel (Bio-Rad). PVDF membranes were activated through a 1 minute 100% ethanol bath and then equilibrated with transfer buffer (3:1:1 ultra-pure water, ethanol 100%, 5X transfer buffer [Bio-Rad]). Proteins were then transferred to activated PDVF membranes (Bio-Rad). Membranes were blocked 1 hour at room temperature with 5% (v/v) non-fat dry milk in TBS with 0.05% (v/v) Tween20 (T- TBS). Membranes were then incubated overnight at 4°C with the appropriate antibodies, listed below. Membranes were then washed with T-TBS and incubated for 1 hour with the appropriate secondary antibody. Signals were revealed using Enhance ChemiLuminescence substrate (ECL Prime) Western Blot detection reagent (GE Healthcare Life Sciences) and the ImageQuant®Las4000Series (GE Healthcare Life Sciences). The relative intensities of the protein bands were quantified using the ImageJ software, and the values were normalized to the intensities of the respective Actin signals.
The following antibodies were used: anti-Actin (1/2000, #MAB1501, Merck Millipore), anti-CRBN (1/1000, ab230411, Abeam), anti-Mcl-1 (1/1000, #5453), anti-Bcl-xL(l/1000, #2764), anti-rabbit polyclonal secondary antibody (1/2000, #7074S) (Cell Signaling Technology) and anti-mouse polyclonal secondary antibody (1/2000, #NA931V-ML, Amersham).
Results
The presence of CRBN was confirmed by western blot in IGROV1-R10 cells as shown in Figure 1.
The effect of the CRBN-PBMl-001 molecule targeting both Mcl-1 and CRBN was characterized in order to demonstrate its ability to target and degrade Mcl-1. Western blot experiments showed that a 24h exposure to Pyridoclax (1 mM) as single agent did not modulate Mcl-1 expression in IGROV1-R10 cells. In contrast, CRBN-PBMl-001 was able to induce a 52% decrease of Mcl-1 expression after a 24h exposure at a concentration of 1 pM as evidenced by densitometry analysis (Figure 2). Moreover, in these experimental conditions, the observed effect was only exerted on Mcl-1, since BCI-XL expression was not affected by the exposure to the CRBN-PBMl-001 and Bcl-2 is not expressed in these cells. These results thus show that CRBN-PBMl-001 is able to bind to Mcl-1 and to initiate its degradation.

Claims (15)

1. A compound of following formula (I) :
Wherein:
Yi, Y2, Y3 and Y4 are each independently C or N, Yi and Y2 being such as one is N and the other is C, Y3 and Y4 being such as one is N and the other is C;
An, An are each independently selected from (C6-Cio)aryl, (5 to 7 membered) heteroaryl, -0-(C6-Cio)aryl, -0-(5 to 7 membered)heteroaryl, -S-(C6-Cio)aryl, -S-(5 to 7 membered)heteroaryl, -(Ci-C6)alkyl-(C6-Cio)aryl, -(Ci-C6)alkyl-(5 to 7 membered) heteroaryl and halogens, in particular Cl, said aryl and heteroaryl groups being optionally substituted by one to three R3 groups, i and j are independently 0 or 1, and
Ri, R2, are at each occurrence, independently selected from (Ci-C6)alkyl, (C6- Cio)aryl, -(Ci-C6)alkyl-(C6-Cio)aryl, -(C2-C6)alkenyl-(C6-Cio)aryl, -carbonyl-(C6-Cio)aryl-, -carbonyl-(Ci-C6)alkyl-(C6-Cio)aryl, C(=0)H, COOH, OH, said alkyl groups being optionally substituted by OH, said aryl groups being optionally substituted by one to three R4 groups; k and 1 are independently 0 or 1 ; at least one of (Ri)k and (R2 being such as:
Ri is selected from (C6-Cio)aryl, -(Ci-C6)alkyl-(C6-Cio)aryl, -(C2-C6)alkenyl- (C6-Cio)aryl, -carbonyl-(C6-Cio)aryl-, -carbonyl-(Ci-C6)alkyl-(C6-Cio)aryl, andk is 1 ; or
R2 is selected from (C6-Cio)aryl, -(Ci-C6)alkyl-(C6-Cio)aryl, -(C2-C6)alkenyl- (C6-Cio)aryl, -carbonyl-(C6-Cio)aryl-, -carbonyl-(Ci-C6)alkyl-(C6-Cio)aryl, and 1 is 1;
R3 IS, at each occurrence, independently selected from (Ci-C6)alkyl, (Ci-C6)alkoxy, OH, C(=0)H, (CH2)mCONRaRb, C(=0)Rc, (CH2)„C02H, (CH2)„C02Rc, (CH2)PCN, (CH2)qC(=N(OH))NH2, -S02NRaRb, I, Cl, Br, F, (C6-Cio)aryl, a (5 to 7 membered)heteroaryl, -(Ci-C6)alkyl-(C6-Cio)aryl, -(C2-C6)alkenyl-(C6-Cio)aryl, said alkyl groups being optionally substituted by a OH, (Ci-C6)alkoxy and/or -0-(C6-Cio)aryl group;
R4 is, at each occurrence, independently selected from (Ci-C6)alkyl, (CH2)nC02H, (CH2)nC02Rc, I, Cl, Br, F, or there are two R4 groups that form with the aryl to which they are bound a naphthyl;
Ra and Rb are independently chosen from H and (C1-C6) alkyl;
Rc is chosen from H and (Ci-C6)alkyl; m is 0, 1,2,3 ; n is 0, 1,2,3 ; p is 0, 1,2,3 ; q is 0, 1,2,3 ; r is 0 or 1 ;
W and Y are each independently selected from a single bond, -CH=CH-, -CºC-, - NH-,-NRs-, -0-, -S-, -C(=0)NH-, -NHC(=0)-, -C(=0)0-, -OC(=0)-;
R5IS, at each occurrence, independently selected from (Ci-C6)alkyl,
Li and L2 are each independently selected from (Ci-Cis)alkyl, (C2-Cis)alkenyl, and (C2-Cis)alkynyl groups, said groups being optionally interrupted by one or more groups selected from -0-, -S-, -NR6-;
Reis, at each occurrence, independently selected from H and (Ci-Ce)alkyl,
X is selected from (5 to 7 membered) heteroaryl, in particular a triazole, -NH- -NR7- , -0-, -S-, -C(=0)NH-, -NHC(=0)-, -C(=0)0-, -0C(=0)-;
R7IS selected from (Ci-Ce)alkyl,
Z is a E3 ubiquitin ligase ligand; or a stereoisomeric form, a mixture of stereoisomeric forms and a pharmaceutically acceptable salt form thereof.
2. The compound according to claim 1, of following formula (II) : (II).
3. The compound according to anyone of the preceding claims, of following formula (Ilia) or (Illb): said compound being in particular of following formula (Ilia 1 ) or (IIIa2), (Illbl) or
(IIIb2):
4. The compound according to anyone of the preceding claims, wherein An and/or An are selected from phenyl, pyridyl, pyrimidyl, imidazolyl, pyrazolyl, thiophenyl, triazolyl, in particular from phenyl, 3 -pyridyl, 5-pyrimidyl, 2-imidazolyl, 3 -pyrazolyl, 2- thiophenyl, 5-triazolyl.
5. The compound according to anyone of the preceding claims, wherein at least one of An, An is a 5 to 7 membered heteroaryl containing a nitrogen atom, preferably pyridyl, notably 3-pyridyl.
6. The compound according to anyone of the preceding claims, wherein Ri and R2 are independently selected from C1-C6 alkyl and (C6-Cio)aryl(C2-C6)alkenyl, preferably from methyl, styryl and 2-(naphthalen-2-yl)vinyl.
7. The compound according to anyone of the preceding claims, wherein Ri is 5- methyl, and/or R2 is 5-styryl or 2-(naphthalen-2-yl)vinyl; or R2 is 5-methyl, and/or Ri is 5- styryl or 2-(naphthalen-2-yl)vinyl
8. The compound according to anyone of the preceding claims, of following formula (IVa) or (IVb): in particular of following formula (IVal), (IVa2), (IVbl) or (IVb2): wherein :
Xi, X2, are at each occurrence, independently selected from C or N;
Ri, R2, R3 are independently, at each occurrence, as defined in any of claims 1 to 7.
9. The compound according to anyone of claim 1 and 2, of one of the following formulae :
wherein :
R3 and R4 are independently, at each occurrence, as defined in any of claims 1 to 7.
10. The compound according to anyone of the preceding claims, wherein: W is -NH-, Li is (Ci-Cis)alkyl, r is 1, X is a (5 to 7 membered) heteroaryl, in particular a triazole, L2 is a (Ci-Cis)alkyl interrupted by one or more -O- atoms, in particular of formula -CH2-(0-CH2-CH2)i-6- and Y is a single bond, -NH- or - C(=0)NH-; or
W is -NH-, Li is a (Ci-Cis)alkyl interrupted by one or more -O- atoms, in particular of formula -CH2-CH2-(0-CH2-CH2)I-6-, r is 0 and Y is a single bond or -C(=0)NH-.
11. The compound according to anyone of claims 1 to 10, wherein Z is selected from:
12. The compound according to claim 1, said compound being selected from:
X is H or Cl.
13. A pharmaceutical composition comprising a compound of formula (I) as defined in anyone of claims 1 to 12, in admixture with at least one pharmaceutically acceptable excipient or carrier.
14. A pharmaceutical composition comprising a compound of formula (I) as defined in anyone of claims 1 to 12, further comprising an additional anti-cancer agent, in particular chosen from anti-EGFR, anti-Map kinases and anti-Akt compounds.
15. A compound of formula (I) according to any of claims 1 to 12, for use in the treatment of cancer, notably of hematologic malignancies, such as lymphoma, leukemia, multiple myeloma, and/or of solid tumors such as ovarian cancers, mesothelioma, melanoma, pancrea, lung, breast, kidney and liver cancers.
AU2021236366A 2020-03-13 2021-03-12 Bcl-2 family proteins modulating compounds for cancer treatment Pending AU2021236366A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP20305263.4 2020-03-13
EP20305263.4A EP3878847A1 (en) 2020-03-13 2020-03-13 Bcl-2 family proteins modulating compounds for cancer treatment
PCT/EP2021/056415 WO2021180966A1 (en) 2020-03-13 2021-03-12 Bcl-2 family proteins modulating compounds for cancer treatment

Publications (1)

Publication Number Publication Date
AU2021236366A1 true AU2021236366A1 (en) 2022-11-03

Family

ID=71120134

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2021236366A Pending AU2021236366A1 (en) 2020-03-13 2021-03-12 Bcl-2 family proteins modulating compounds for cancer treatment

Country Status (6)

Country Link
EP (2) EP3878847A1 (en)
JP (1) JP2023516790A (en)
CN (1) CN115715286A (en)
AU (1) AU2021236366A1 (en)
CA (1) CA3170893A1 (en)
WO (1) WO2021180966A1 (en)

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2915811A1 (en) 2014-03-04 2015-09-09 Centre Régional de Lutte contre le Cancer - Centre François Baclesse MCL-1 modulating compounds for cancer treatment
IL295603B2 (en) * 2017-09-22 2024-03-01 Kymera Therapeutics Inc Protein degraders and uses thereof
EP3737666A4 (en) * 2018-01-12 2022-01-05 Kymera Therapeutics, Inc. Protein degraders and uses thereof
US20220348556A1 (en) * 2018-07-06 2022-11-03 Kymera Therapeutics, Inc. Protein degraders and uses thereof

Also Published As

Publication number Publication date
EP4118080A1 (en) 2023-01-18
EP3878847A1 (en) 2021-09-15
WO2021180966A1 (en) 2021-09-16
EP4118080B1 (en) 2024-02-21
CN115715286A (en) 2023-02-24
EP4118080C0 (en) 2024-02-21
CA3170893A1 (en) 2021-09-16
JP2023516790A (en) 2023-04-20

Similar Documents

Publication Publication Date Title
JP7208285B2 (en) 1,4-disubstituted pyridazine derivatives and their use to treat conditions associated with SMN deficiency
CA3030510C (en) Azole dione compounds with anti-cancer activity
DK2473504T3 (en) Heterocyclic Compounds for Inhibition of Passover
US20110053931A1 (en) Quinoline compounds and methods of use
CA3035860A1 (en) Pyridine compound
JP5898230B2 (en) Novel substituted bicyclic aromatic compounds as S-nitrosoglutathione reductase inhibitors
EP2185521A2 (en) Compounds and methods for modulating rho gtpases
CN113966324A (en) NOTCH signaling pathway inhibitors and their use in cancer treatment
Pati et al. Novel metal chelators thiosemicarbazones with activity at the σ 2 receptors and P-glycoprotein: an innovative strategy for resistant tumor treatment
EP3114119B1 (en) Mcl-1 modulating compounds for cancer treatment
JP7102388B2 (en) Monocyclic heteroaryl substituted compound
EP4118080B1 (en) Bcl-2 family proteins modulating compounds for cancer treatment
CN116348114A (en) Thiobenzimidazole derivatives or pharmaceutically acceptable salts thereof and use thereof
KR20220068224A (en) PERK inhibitory compounds
WO2022166990A1 (en) Anti-tumor pharmaceutical combination
WO2023016511A1 (en) Azaaryl compound, and preparation method therefor and use thereof
KR20220051816A (en) Thiobenzimidazole derivatives or its pharmaceutically acceptable salts and use thereof